PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 23279643-0 2013 The VKORC1 and CYP2C9 genotypes are associated with over-anticoagulation during initiation of warfarin therapy in children. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 23348161-0 2013 The relationship between CYP2C9 gene polymorphisms and upper gastrointestinal bleeding in patients who used warfarin. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 23348161-3 2013 CYP2C9 is a hepatic microsomal enzyme and has a primary role in metabolism of warfarin and genetic variations of CYP2C9 may cause a serious effect on the response to warfarin in patients. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23348161-3 2013 CYP2C9 is a hepatic microsomal enzyme and has a primary role in metabolism of warfarin and genetic variations of CYP2C9 may cause a serious effect on the response to warfarin in patients. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23348161-3 2013 CYP2C9 is a hepatic microsomal enzyme and has a primary role in metabolism of warfarin and genetic variations of CYP2C9 may cause a serious effect on the response to warfarin in patients. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 23348161-4 2013 The aim of this study was to determine the efficiency of CYP2C9 gene polymorphisms on drug metabolism in patients who had upper gastrointestinal system bleeding while using warfarin. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 23348161-11 2013 CONCLUSION: CYP2C9*2 and CYP2C9*3 polymorphisms were related to the increase of excessive anticoagulation and bleeding risk in the patients who used warfarin. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 23348161-11 2013 CONCLUSION: CYP2C9*2 and CYP2C9*3 polymorphisms were related to the increase of excessive anticoagulation and bleeding risk in the patients who used warfarin. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 23452606-2 2013 Although numerous candidate gene studies in epilepsy PGX have been published, to date only two validated associations exist: the association of the *2 and *3 alleles of CYP2C9 with phenytoin metabolism and the association of HLA-B*1502 with serious hypersensitivity reactions to carbamazepine. Phenytoin 181-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 23241944-3 2013 RESULTS: The rate of generation of human-predominant drug metabolites for S-mephenytoin and diclofenac in the chimeric mice was correlated with the CYP2C19 (n=9 donors, P=0.0005) or CYP2C9 (n=7 donors, P=0.0394) genotype, respectively, of the transplanted human hepatocytes. Mephenytoin 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 23241944-3 2013 RESULTS: The rate of generation of human-predominant drug metabolites for S-mephenytoin and diclofenac in the chimeric mice was correlated with the CYP2C19 (n=9 donors, P=0.0005) or CYP2C9 (n=7 donors, P=0.0394) genotype, respectively, of the transplanted human hepatocytes. Diclofenac 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 23469684-0 2013 Simvastatin pharmacokinetics in healthy Chinese subjects and its relations with CYP2C9, CYP3A5, ABCB1, ABCG2 and SLCO1B1 polymorphisms. Simvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 23159229-0 2013 The influence of VKORC1 and CYP2C9 gene sequence variants on the stability of maintenance phase warfarin treatment. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 23159229-1 2013 INTRODUCTION: A substantial part of the inter-individual variation in vitamin K-antagonist dose can be explained by certain sequence variants in the genes CYP2C9 (NG_008385.1:g.8633C>T or *1/*2, NG_008385.1:g.47639A>C or *1/*3) and VKORC1 (NG_011564.1:g.6399C>T). Vitamin K 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 23159229-7 2013 CONCLUSIONS: We observed modest effects of common gene sequence variants in CYP2C9 and VKORC1 on stability of maintenance phase warfarin therapy. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 23640947-6 2013 Among genes associated with metabolism of the most commonly applied anaesthetics such as propofol and sevoflurane, the following ones can be mentioned: CYP2E1, CYP2B6, CYP2C9, GSTP1, UGT1A9, SULT1A1 and NQO1. Propofol 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 23640947-6 2013 Among genes associated with metabolism of the most commonly applied anaesthetics such as propofol and sevoflurane, the following ones can be mentioned: CYP2E1, CYP2B6, CYP2C9, GSTP1, UGT1A9, SULT1A1 and NQO1. Sevoflurane 102-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 23118328-3 2013 The aim of this study is to evaluate the possible association between sex, polymorphisms in the CYP2C8 and CYP2C9 genes, and the pharmacokinetics of losartan, valsartan, candesartan, and telmisartan. Telmisartan 187-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 23302640-6 2013 In a metabolic inhibition study using human liver microsomes, cytochrome P450 (CYP) 2C9-mediated hydroxylation of S-warfarin was almost unaffected by MTZ at the corresponding concentrations. Warfarin 114-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-87 23774101-0 2013 The bleeding risk during warfarin therapy is associated with the number of variant alleles of CYP2C9 and VKORC1 genes. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 23774101-4 2013 AIM: It was the aim of this study to assess the impact of the total variant allele count of CYP2C9 and VKORC1 genes on bleeding related to warfarin treatment. Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 22917719-0 2013 A multiplex assay to detect variations in the CYP2C9, VKORC1, CYP4F2 and APOE genes involved in acenocoumarol metabolism. Acenocoumarol 96-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 22917719-3 2013 DESIGN AND METHODS: We developed a PCR multiplex SNaPshot reaction that targets 6 SNPs (single nucleotide polymorphisms) in CYP2C9, CYP4F2, VKORC1 and APOE genes, which are associated with acenocoumarol dose requirements. Acenocoumarol 189-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 23118328-3 2013 The aim of this study is to evaluate the possible association between sex, polymorphisms in the CYP2C8 and CYP2C9 genes, and the pharmacokinetics of losartan, valsartan, candesartan, and telmisartan. Losartan 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 22892446-1 2013 The aim of this study was to reveal the contribution of CYP4F2, CYP2C9, and VKORC1 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of warfarin in Japanese pediatric patients. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 22892446-6 2013 In patients with the CYP2C9 *3 allele, the unbound oral clearance values (normalized to body surface area) for S-warfarin were found to be significantly lower than in patients with the wild-type allele. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 23118328-3 2013 The aim of this study is to evaluate the possible association between sex, polymorphisms in the CYP2C8 and CYP2C9 genes, and the pharmacokinetics of losartan, valsartan, candesartan, and telmisartan. Valsartan 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 23118328-7 2013 CYP2C9 variants were associated only with losartan pharmacokinetics: the half-life of losartan was higher in CYP2C9*3 allele carriers (3.1 +- 0.4 hours) than in volunteers with the wild-type genotype (2.3 +- 0.1 hours) (P <= 0.05). Losartan 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23229783-2 2013 Recently, we reported that culture of human hepatocellular carcinoma FLC-4 cells on micro-space cell culture plates could increase the functional expression of drug-metabolizing enzymes including CYP3A4 and CYP2C9, which are involved in 1"-hydroxylation and 6-hydroxylation of BBR, respectively. Benzbromarone 277-280 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 23118328-7 2013 CYP2C9 variants were associated only with losartan pharmacokinetics: the half-life of losartan was higher in CYP2C9*3 allele carriers (3.1 +- 0.4 hours) than in volunteers with the wild-type genotype (2.3 +- 0.1 hours) (P <= 0.05). Losartan 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 23118328-7 2013 CYP2C9 variants were associated only with losartan pharmacokinetics: the half-life of losartan was higher in CYP2C9*3 allele carriers (3.1 +- 0.4 hours) than in volunteers with the wild-type genotype (2.3 +- 0.1 hours) (P <= 0.05). Losartan 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23118328-7 2013 CYP2C9 variants were associated only with losartan pharmacokinetics: the half-life of losartan was higher in CYP2C9*3 allele carriers (3.1 +- 0.4 hours) than in volunteers with the wild-type genotype (2.3 +- 0.1 hours) (P <= 0.05). Losartan 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 23257392-1 2013 The cytochrome P450 (P450, CYP) 2A6 inhibitor chalepensin was found to inhibit human CYP1A1, CYP1A2, CYP2A13, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 to different extents. 3-(alpha,alpha-dimethylallyl)psoralen 46-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 23385314-3 2013 Efavirenz was a moderate inhibitor of CYP2C9 (K(i) = 19.46 microM) and CYP2C19 (K(i) = 21.31 microM); and a weak inhibitor of CYP3A (K(i) = 40.33 microM). efavirenz 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 23118328-9 2013 These results suggest that genotypes for CYP2C9 and CYP2C8 are relevant to the pharmacokinetics of losartan and valsartan, respectively, but not the pharmacokinetics of candesartan or telmisartan. Losartan 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 23118328-9 2013 These results suggest that genotypes for CYP2C9 and CYP2C8 are relevant to the pharmacokinetics of losartan and valsartan, respectively, but not the pharmacokinetics of candesartan or telmisartan. Valsartan 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 23481074-0 2013 Cytochrome P450 (CYP2C9*2,*3) & vitamin-K epoxide reductase complex (VKORC1 -1639G<A) gene polymorphisms & their effect on acenocoumarol dose in patients with mechanical heart valve replacement. Acenocoumarol 134-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 23545593-0 2013 A case-report of unpredictable and massive voriconazole intoxication in a patient with extensive CYP2C19 and CYP2C9 polymorphisms. Voriconazole 43-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 23545593-1 2013 This case-report describes a massive voriconazole (VRZ) intoxication in a patient with a poor metabolizer profile, highlighted by low plasma main metabolite concentrations (N-oxide voriconazole), despite an extensive genetic profile for CYP2C19 and CYP2C9. Voriconazole 37-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 23545593-1 2013 This case-report describes a massive voriconazole (VRZ) intoxication in a patient with a poor metabolizer profile, highlighted by low plasma main metabolite concentrations (N-oxide voriconazole), despite an extensive genetic profile for CYP2C19 and CYP2C9. Voriconazole 51-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 23481074-1 2013 BACKGROUND & OBJECTIVES: Studies have demonstrated the effect of CYP2C9 (cytochrome P450) and VKORC1 (vitamin K epoxide reductase complex) gene polymorphisms on the dose of acenocoumarol. Adenosine Monophosphate 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 23481074-1 2013 BACKGROUND & OBJECTIVES: Studies have demonstrated the effect of CYP2C9 (cytochrome P450) and VKORC1 (vitamin K epoxide reductase complex) gene polymorphisms on the dose of acenocoumarol. Acenocoumarol 177-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 23481074-3 2013 The aim of this study was to determine the occurrence of CYP2C9*2,*3 and VKORC 1 -1639G>A gene polymorphisms and to study their effects on the dose of acenocoumarol required to maintain a target International Normalized Ratio (INR) in patients with mechanical heart valve replacement. Acenocoumarol 154-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 23481074-12 2013 INTERPRETATION & CONCLUSIONS: The study findings point towards the role of CYP2C9 and VKORC1 gene polymorphisms in determining the inter-individual dose variability of acenocoumarol in the Indian patients with mechanical heart valve replacement. Acenocoumarol 172-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 24324494-3 2013 The gene variants in CYP2C9, ABCC8/KCNJ11, and TCF7L2 were associated with the effect of sulfonylureas. Sulfonylurea Compounds 89-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24324494-4 2013 CYP2C9 encodes sulfonylurea metabolizing cytochrome P450 isoenzyme 2C9, ABCC8 and KCNJ11 genes encode proteins constituting ATP-sensitive K(+) channel which is a therapeutic target for sulfonylureas, and TCF7L2 is a gene with the strongest association with type 2 diabetes. Sulfonylurea Compounds 15-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24324494-4 2013 CYP2C9 encodes sulfonylurea metabolizing cytochrome P450 isoenzyme 2C9, ABCC8 and KCNJ11 genes encode proteins constituting ATP-sensitive K(+) channel which is a therapeutic target for sulfonylureas, and TCF7L2 is a gene with the strongest association with type 2 diabetes. Sulfonylurea Compounds 185-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23958198-3 2013 Based on in vitro evidence cranberry juice is an inhibitor of CYP enzymes and at higher amounts as potent as ketoconazole (CYP3A) and fluconazole (CYP2C9). Fluconazole 134-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 23400742-1 2013 The nonnucleoside reverse transcriptase inhibitor etravirine, approved for use in treatment-experienced, HIV-1-infected patients, is a substrate and inducer of cytochrome P450 (CYP) 3A4 and a substrate and inhibitor of CYP2C9/CYP2C19. etravirine 50-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 23563037-0 2013 Variability in CYP2C9 allele frequency: a pilot study of its predicted impact on warfarin response among healthy South and North Indians. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 24800477-0 2013 [Relationship between warfarin dosing and activity of CYP2C9 assessed by the content of losartan and its metabolite E-3174 in the urine of patients with mechanical prosthetic heart valves]. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24800477-0 2013 [Relationship between warfarin dosing and activity of CYP2C9 assessed by the content of losartan and its metabolite E-3174 in the urine of patients with mechanical prosthetic heart valves]. Losartan 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24800477-0 2013 [Relationship between warfarin dosing and activity of CYP2C9 assessed by the content of losartan and its metabolite E-3174 in the urine of patients with mechanical prosthetic heart valves]. losartan carboxylic acid 116-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24800477-1 2013 We evaluated effect of activity of cytochrome P450 CYP2C9 on maintenance doses of warfarin in 33 patients with implanted artificial heart valves. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 24858991-1 2013 BACKGROUND AND OBJECTIVE: VKORC1, CYP2C9, and CYP4F2 are known to be responsible for the metabolism of warfarin. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 23111422-1 2013 OBJECTIVES: Etravirine (ETV) is metabolized by cytochrome P450 (CYP) 3A, 2C9, and 2C19. etravirine 12-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-76 23111422-1 2013 OBJECTIVES: Etravirine (ETV) is metabolized by cytochrome P450 (CYP) 3A, 2C9, and 2C19. etravirine 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-76 23343733-2 2013 Used concomitantly with coumarin derivatives, it may enhance their anticoagulant effects by inhibiting the CYP2C9 isoenzyme; an interaction which has been well described for oral and vaginal administrations of miconazole. coumarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 23343733-2 2013 Used concomitantly with coumarin derivatives, it may enhance their anticoagulant effects by inhibiting the CYP2C9 isoenzyme; an interaction which has been well described for oral and vaginal administrations of miconazole. Miconazole 210-220 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 24399734-0 2013 Effect of CYP2C9 and VKORC1 genetic variations on warfarin dose requirements in Indian patients. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 24399734-2 2013 The management of warfarin therapy is challenging because it shows large inter and intra individual variability in patient response due to factors like age, gender, diet, concurrent drug interactions and variations in CYP2C9 and VKORC1 genes. Warfarin 18-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 24399734-3 2013 Studies implicate that polymorphisms in VKORC1 and CYP2C9 genes are associated with reduced doses of warfarin. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 24399734-4 2013 The aim of our current study was to characterize the effects of VKORC1 and CYP2C9 gene variations that contribute to variability in warfarin dosing in Indian patients. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 23563037-8 2013 The variation in the frequency of combined CYP2C9/ VKORC1 genotypes revealed plausible difference in warfarin response among SI and NI. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 23196355-7 2013 In conclusion, we developed a prediction algorithm using clinical data and three polymorphisms in VKORC1, CYP2C9* and CYP4F2 genes that provided a steady acenocoumarol dose for about 50% of patients in the Validation Cohort. Acenocoumarol 158-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 22917556-6 2012 Formation of 12-hydroxyellipticine is generated mainly by CYP2C19, followed by CYP2C9>3A4>2D6>2E1 and >2A6. 12-hydroxyellipticine 13-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 23237631-6 2012 The aim of this study is to evaluate the effectiveness and efficiency of an acenocoumarol dosing algorithm developed by our group which includes demographic, clinical and pharmacogenetic variables (VKORC1, CYP2C9, CYP4F2 and ApoE) in patients with venous thromboembolism (VTE). Acenocoumarol 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 23089473-7 2012 Hypoxia induced SP cell proliferation, and microarray analysis showed that the expression of CYP2C9 was significantly increased in hypoxic than normoxic SP cells. sp 16-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 23342320-0 2012 CYP2C9 Mutation Affecting the Individual Variability of Warfarin Dose Requirement. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23342320-5 2012 There are two identified genes that are responsible for the main proportion of the genetic effect: CYP2C9, which codes for the enzyme cytochrome P450 2C9 that metabolizes S-warfarin, and VKORC1, which codes for warfarin"s target, vitamin K epoxide reductase. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 23342320-5 2012 There are two identified genes that are responsible for the main proportion of the genetic effect: CYP2C9, which codes for the enzyme cytochrome P450 2C9 that metabolizes S-warfarin, and VKORC1, which codes for warfarin"s target, vitamin K epoxide reductase. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-153 23342320-5 2012 There are two identified genes that are responsible for the main proportion of the genetic effect: CYP2C9, which codes for the enzyme cytochrome P450 2C9 that metabolizes S-warfarin, and VKORC1, which codes for warfarin"s target, vitamin K epoxide reductase. Warfarin 211-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 23342320-5 2012 There are two identified genes that are responsible for the main proportion of the genetic effect: CYP2C9, which codes for the enzyme cytochrome P450 2C9 that metabolizes S-warfarin, and VKORC1, which codes for warfarin"s target, vitamin K epoxide reductase. Warfarin 211-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-153 23342320-6 2012 We report a case of intolerance to warfarin dosing, due to impaired drug metabolism in a patient with CYP2C9(*)1/(*)3 and VKORC 1173TT. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 23089473-9 2012 These results indicate that STAT3 is critical for CSC survival and hypoxia-inducible CYP2C9 expression is responsible the doxorubicin resistance of CSCs under hypoxic conditions. Doxorubicin 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 23132553-7 2012 Our results show that the CYP4F2 p.V433M polymorphism is associated with interindividual variability in response to coumarin drugs, but with a low effect size that is confirmed to be lower than those contributed by VKORC1 and CYP2C9 polymorphisms. coumarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 226-232 23018470-1 2012 BACKGROUND AND OBJECTIVE: Genetic polymorphisms of cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) and patient demographic characteristics are responsible for inter-individual differences in warfarin maintenance dosage requirements. Warfarin 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-76 23781667-3 2012 The variability of Warfarin dosage is largely genetically determined, and it can be partly explained by the C1173T and G-1639A polymorphisms of vitamin K epoxide reductase complex subunit 1 (VKORC1) which is its target and *2 and *3 allele of Cytochrome P-450 (CYP) 2C9 [CYP2C9] enzyme which metabolizes to its inactive form. Warfarin 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 243-269 23018470-1 2012 BACKGROUND AND OBJECTIVE: Genetic polymorphisms of cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) and patient demographic characteristics are responsible for inter-individual differences in warfarin maintenance dosage requirements. Warfarin 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 23153186-2 2012 AREAS COVERED: This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. Sulfonylurea Compounds 83-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 23153186-2 2012 AREAS COVERED: This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. Sulfonylurea Compounds 99-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 23153186-2 2012 AREAS COVERED: This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. meglitinide 105-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 23153186-2 2012 AREAS COVERED: This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. Thiazolidinediones 123-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 23153186-2 2012 AREAS COVERED: This review focuses on the effect of CYP2C enzymes on metabolism of sulphonylureas (SUs), meglitinides, and thiazolidinediones (TZDs) discussing their impact on pharmacokinetics, drug interactions and toxicological profiles. Thiazolidinediones 143-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 23153186-4 2012 EXPERT OPINION: Genetic polymorphisms of CYP2C9 enzymes (*2/*2, *2/*3, *3/*3) influence the glycaemic response to SUs and impair their substrate metabolism. Sulfonylurea Compounds 114-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 23153186-6 2012 Functional polymorphisms of CYP2C8- and CYP2C9 drug metabolizing genes affect markedly pharmacokinetics of meglitinides. meglitinide 107-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 23781667-0 2012 Prevalence of VKORC1 and CYP2C9 gene polymorphisms in Indian population and its effect on warfarin response. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 23781667-3 2012 The variability of Warfarin dosage is largely genetically determined, and it can be partly explained by the C1173T and G-1639A polymorphisms of vitamin K epoxide reductase complex subunit 1 (VKORC1) which is its target and *2 and *3 allele of Cytochrome P-450 (CYP) 2C9 [CYP2C9] enzyme which metabolizes to its inactive form. Warfarin 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 23781667-14 2012 CONCLUSION: In the present study the effect of CYP2C9*2, *3, and VKORC1 (C1173T and G-1639A) genotypes on warfarin dose was observed. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 22205719-2 2012 Bosentan, an endothelin (ET) receptor-type ET(A) and ET(B) antagonist, is metabolized to active metabolites by and an inducer of cytochrome P450 (CYP)2C9 and CYP3A. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-153 23104259-10 2012 CYP2C9*3 may be the main genetic factor in hemorrhagic complications in Chinese patients under warfarin anticoagulation. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23089684-9 2012 Our data set included CYP2C9*3 (rs1057910) and CYP2C19*2 (rs4244285) enzyme activity-altering variants associated in a recent genome-wide study with acenocoumarol-induced and warfarin-induced anticoagulation or to the antiplatelet effect of clopidogrel, respectively. Acenocoumarol 149-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 23015069-8 2012 In conclusion, PG refinement algorithm integrating early INR response and three genotypes (CYP2C9*3, VKORC1-1639A/G, CYP4F2 rs2108622) improves the accuracy of warfarin dose prediction in Chinese patients mainly under low-intensity anticoagulation. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 23089684-14 2012 However, besides the activity-altering CYP2C9*3 (rs1057910) and CYP2C19*2 (rs4244285) variants, the high derived allele frequencies of novel recently identified acenocoumarol genome-wide associated SNPs at CYP2C9 (rs4086116) and CYP2C18 (rs12772169, rs1998591, rs2104543, rs1042194) loci variants indicated that the CYP2C region may have been influenced by selection. Acenocoumarol 161-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 22259022-9 2012 In order to investigate whether the polymorphism of CYP2C9 and CYP2C19 enzymes could be related to the metabolism of mitotane, RT-PCR analysis was performed. Mitotane 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. Hydrogen 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. Camphor 30-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. Hydrogen 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. cyclohexene 84-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. propylene 100-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 26605619-5 2012 The hydrogen abstraction from camphor, and hydrogen abstraction and C-O addition of cyclohexene and propene by P450cam have been modeled, along with the addition of benzene to Compound I in CYP2C9, at the B3LYP-D2/CHARMM27 level of theory. Benzene 165-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 22941809-0 2012 CYP2C9*3(1075A>C), MDR1 G2677T/A and MDR1 C3435T are determinants of inter-subject variability in fluvastatin pharmacokinetics in healthy Chinese volunteers. Fluvastatin 101-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 22941809-1 2012 To evaluate the impact of single-nucleotide polymorphisms (SNPs) in CYP2C9, MDR1, SLCO1B1 and ABCG2 on the pharmacokinetics of fluvastatin in Chinese participants.A pharmacokinetic study of fluvastatin (single dose 40 mg) was conducted in 12 healthy Chinese volunteers. single-nucleotide 26-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 22941809-1 2012 To evaluate the impact of single-nucleotide polymorphisms (SNPs) in CYP2C9, MDR1, SLCO1B1 and ABCG2 on the pharmacokinetics of fluvastatin in Chinese participants.A pharmacokinetic study of fluvastatin (single dose 40 mg) was conducted in 12 healthy Chinese volunteers. Fluvastatin 127-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 22941809-4 2012 The SNPs were determined by TaqMan (MGB) genotyping assay.Effect of CYP2C9*3 (c.1075A>C) on area under the plasma concentration-time curve (AUC) of fluvastatin was statistically significant. Fluvastatin 151-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 23287317-3 2012 Polymorphic alleles CYP2C9 FNx01 2 and CYP2C9 FNx01 3 occur in the Indian population and this may serve as the basis for using genotyping as a tool to predict phenytoin toxicity. Phenytoin 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 22791347-3 2012 The present study reviews the literature on genetic contributions to fluoxetine response in children and adults, and concludes that the different polymorphisms of CYP2D6 and CYP2C9 may influence the blood concentrations of fluoxetine. Fluoxetine 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 22791347-3 2012 The present study reviews the literature on genetic contributions to fluoxetine response in children and adults, and concludes that the different polymorphisms of CYP2D6 and CYP2C9 may influence the blood concentrations of fluoxetine. Fluoxetine 223-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 23287317-0 2012 Association of CYP2C9 polymorphisms with phenytoin toxicity in Indian patients. Phenytoin 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 22933344-10 2012 The formation of 5,6-dihydroxy BBR from 6-hydroxy BBR was catalysed by recombinant CYP2C9 and CYP1A2. 5,6-dihydroxy bbr 17-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 22933344-10 2012 The formation of 5,6-dihydroxy BBR from 6-hydroxy BBR was catalysed by recombinant CYP2C9 and CYP1A2. 6-hydroxybenzbromarone 40-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 1'-hydroxybenzbromarone 48-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 1'-hydroxybenzbromarone 48-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 6-hydroxybenzbromarone 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 6-hydroxybenzbromarone 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 5,6-dihydroxy bbr 163-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 22933344-13 2012 These results suggest that (i) the formation of 1"-hydroxy BBR and 6-hydroxy BBR is mainly catalysed by CYP3A4 and CYP2C9, respectively, and (ii) the formation of 5,6-dihydroxy BBR is catalysed by CYP2C9 and CYP1A2 in human liver microsomes. 5,6-dihydroxy bbr 163-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 22896727-1 2012 (S)-Warfarin 7-hydroxylation and midazolam 1"-hydroxylation are among the preferred probe substrate reactions for CYP2C9 and CYP3A4/5, respectively. Warfarin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 22896727-1 2012 (S)-Warfarin 7-hydroxylation and midazolam 1"-hydroxylation are among the preferred probe substrate reactions for CYP2C9 and CYP3A4/5, respectively. Midazolam 33-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 22859722-3 2012 Raloxifene, a drug known to undergo CYP3A-mediated reactive metabolite formation and time-dependent inhibition in vitro, was used to explore the potential for bioactivation and enzyme inactivation of additional P450 enzymes (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A5). Raloxifene Hydrochloride 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 241-247 23287317-3 2012 Polymorphic alleles CYP2C9 FNx01 2 and CYP2C9 FNx01 3 occur in the Indian population and this may serve as the basis for using genotyping as a tool to predict phenytoin toxicity. Phenytoin 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 23287317-4 2012 AIMS: To evaluate the association between the presence of polymorphic alleles CYP2C9 FNx01 2 and FNx013 and phenytoin toxicity in Indian patients with epilepsy. Phenytoin 108-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 23287317-16 2012 We found a significant association between polymorphic alleles CYP2C9 FNx01 2 and FNx013 and toxic phenytoin levels. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 22853867-4 2012 RESULTS: The AUC and t1/2 of glibenclamide following the microdosing tended to be higher and longer, respectively, in CYP2C9*1/*3 than CYP2C9*1/*1 subjects. Glyburide 29-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 23148630-0 2012 Influence of the CYP2C9*3 allele on the pharmacological interaction between warfarin and simvastatin. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 23148630-0 2012 Influence of the CYP2C9*3 allele on the pharmacological interaction between warfarin and simvastatin. Simvastatin 89-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 23148630-2 2012 A clinically significant interaction between warfarin and simvastatin is unique to carriers of the CYP2C9*3 allele. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 23041981-0 2012 Effect of genetic variants, especially CYP2C9 and VKORC1, on the pharmacology of warfarin. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 23041981-1 2012 The genes encoding the cytochrome P450 2C9 enzyme (CYP2C9) and vitamin K-epoxide reductase complex unit 1 (VKORC1) are major determinants of anticoagulant response to warfarin. Warfarin 167-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-42 23041981-1 2012 The genes encoding the cytochrome P450 2C9 enzyme (CYP2C9) and vitamin K-epoxide reductase complex unit 1 (VKORC1) are major determinants of anticoagulant response to warfarin. Warfarin 167-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 22571484-4 2012 In addition, it has been proposed that the residues involved in substrate recognition sites (SRS) 1, SRS 3, and/or SRS 4 are important for the metabolizing capacity and/or the substrate binding of CYP2C9 and CYP2C19. srs 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 22533669-4 2012 SYBR Green-based real-time polymerase chain reaction (PCR) assay was used for studying VKORC1 (C1173T) and CYP2C9*3 polymorphisms. sybr green 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 22533669-8 2012 VKORC1 TT and CYP2C9*2\*2 were associated with a significantly lower warfarin dose. Warfarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 22533669-9 2012 VKORC1 and CYP2C9 accounted for 31.7% and 15.6% of warfarin dose variability, respectively, and together with clinical factors explained 61.3% of total variability. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 23148630-2 2012 A clinically significant interaction between warfarin and simvastatin is unique to carriers of the CYP2C9*3 allele. Simvastatin 58-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 23113310-0 2012 CYP2C9 and VKORC1 gene polymorphism is inessential for bleeding development under conditions of oral application of anticoagulant acenocoumarol in Russian patients at high risk of thromboembolic complications. Acenocoumarol 130-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 22752007-4 2012 In addition, we revealed that sesamin caused a mechanism-based inhibition (MBI) of CYP2C9, the P450 enzyme responsible for sesamin monocatecholization. sesamin 30-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 22752007-4 2012 In addition, we revealed that sesamin caused a mechanism-based inhibition (MBI) of CYP2C9, the P450 enzyme responsible for sesamin monocatecholization. sesamin 123-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 22752007-7 2012 The P450 enzymes responsible for S- and R-monocatechol formation were CYP2C9 and CYP1A2, respectively. Sulfur 33-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 22752007-7 2012 The P450 enzymes responsible for S- and R-monocatechol formation were CYP2C9 and CYP1A2, respectively. r-monocatechol 40-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 22853867-1 2012 OBJECTIVES: To test whether the multiple phenotype and genotype relationships established using therapeutic dose, can be reproduced following oral microdosing using substrates of CYP2C9 (warfarin and glibenclamide), CYP2C19 (lansoprazole), CYP2D6 (dextromethorphan), and OATPs (glibenclamide). Warfarin 187-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 22853867-4 2012 RESULTS: The AUC and t1/2 of glibenclamide following the microdosing tended to be higher and longer, respectively, in CYP2C9*1/*3 than CYP2C9*1/*1 subjects. Glyburide 29-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Chlorpropamide 296-310 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 22486182-9 2012 A final multivariate regression model that included the target International Normalized Ratio, VKORC1 and CYP2C9 genotypes explained 36 2% of the overall interindividual variability in acenocoumarol dose requirement. Acenocoumarol 185-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 22486182-11 2012 VKORC1 and CYP2C9 variants significantly affected acenocoumarol dose, in-line with results in other populations. Acenocoumarol 50-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 22486182-12 2012 For the Moroccan population, the SNPs that have the largest effect on acecoumarol dose are CYP2C9 rs1799853, CYP2C9 rs1057910 and VKORC1 rs9923231. acecoumarol 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 22486182-12 2012 For the Moroccan population, the SNPs that have the largest effect on acecoumarol dose are CYP2C9 rs1799853, CYP2C9 rs1057910 and VKORC1 rs9923231. acecoumarol 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 22943175-9 2012 The inhibitory action of glycyrrhetinic acid was observed in CYP2C9 for 4-hydroxylation of diclofenac, CYP2C19 for 4"-hydroxylation of (S)-mephenytoin and CYP3A4 for 1"-hydroxylation of midazolam with half maximal inhibitory concentration (IC50) values of 4.3-fold, 3.8-fold and 9.6-fold higher than specific inhibitors in HLM, respectively. Glycyrrhetinic Acid 25-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 22943175-9 2012 The inhibitory action of glycyrrhetinic acid was observed in CYP2C9 for 4-hydroxylation of diclofenac, CYP2C19 for 4"-hydroxylation of (S)-mephenytoin and CYP3A4 for 1"-hydroxylation of midazolam with half maximal inhibitory concentration (IC50) values of 4.3-fold, 3.8-fold and 9.6-fold higher than specific inhibitors in HLM, respectively. Diclofenac 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 22943175-9 2012 The inhibitory action of glycyrrhetinic acid was observed in CYP2C9 for 4-hydroxylation of diclofenac, CYP2C19 for 4"-hydroxylation of (S)-mephenytoin and CYP3A4 for 1"-hydroxylation of midazolam with half maximal inhibitory concentration (IC50) values of 4.3-fold, 3.8-fold and 9.6-fold higher than specific inhibitors in HLM, respectively. Mephenytoin 135-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 23047421-2 2012 Genotypes for two single nucleotide polymorphisms (SNPs) in the cytochrome P 450 2C9 gene, affecting warfarin metabolism, and one SNP in vitamin K reductase complex 1 gene, affecting warfarin sensitivity, account for approximately 30% of therapeutic warfarin dosing variability in whites and Asians. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-84 23047421-2 2012 Genotypes for two single nucleotide polymorphisms (SNPs) in the cytochrome P 450 2C9 gene, affecting warfarin metabolism, and one SNP in vitamin K reductase complex 1 gene, affecting warfarin sensitivity, account for approximately 30% of therapeutic warfarin dosing variability in whites and Asians. Warfarin 183-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-84 23047421-2 2012 Genotypes for two single nucleotide polymorphisms (SNPs) in the cytochrome P 450 2C9 gene, affecting warfarin metabolism, and one SNP in vitamin K reductase complex 1 gene, affecting warfarin sensitivity, account for approximately 30% of therapeutic warfarin dosing variability in whites and Asians. Warfarin 183-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-84 22947619-6 2012 The surrounding SAM, a polyethylene glycol silane, displayed very little affinity toward the P450 isozyme CYP2C9, as demonstrated by x-ray photoelectron spectroscopy and surface plasmon resonance. poly(ethylene glycol)silane 23-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Phenytoin 269-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Phenytoin 269-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Phenytoin 269-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Phenytoin 269-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Tolbutamide 280-291 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Tolbutamide 280-291 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Tolbutamide 280-291 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Tolbutamide 280-291 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22854539-2 2012 We studied the functional polymorphisms of CYP2C9, CYP4F2 and VKORC1 genes to evaluate their impact on the warfarin maintenance dose in an admixed Omani patient cohort with Caucasian, African and Asian ancestries. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 22854539-4 2012 Univariate analysis revealed that age, gender, weight, atrial fibrillation, deep vein thrombosis/pulmonary embolism and variant genotypes of CYP2C9 and VKORC1 loci were significantly associated with warfarin dose in the studied patient population. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 22943175-12 2012 The results suggest that glycyrrhetinic acid has the potential to interact with a wide range of xenobiotics or endogenous chemicals that are CYP2C9, CYP2C19 and CYP3A4 substrates. Glycyrrhetinic Acid 25-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 23156991-5 2012 The results revealed that a combination of glimepiride with piperine led to the enhancement of the bioavailability of glimepiride by inhibiting the CYP2C9 enzyme, which suggested that piperine might be beneficial as an adjuvant to glimepiride in a proper dose, in diabetic patients. glimepiride 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 23156991-5 2012 The results revealed that a combination of glimepiride with piperine led to the enhancement of the bioavailability of glimepiride by inhibiting the CYP2C9 enzyme, which suggested that piperine might be beneficial as an adjuvant to glimepiride in a proper dose, in diabetic patients. piperine 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 23156991-5 2012 The results revealed that a combination of glimepiride with piperine led to the enhancement of the bioavailability of glimepiride by inhibiting the CYP2C9 enzyme, which suggested that piperine might be beneficial as an adjuvant to glimepiride in a proper dose, in diabetic patients. glimepiride 118-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 23156991-5 2012 The results revealed that a combination of glimepiride with piperine led to the enhancement of the bioavailability of glimepiride by inhibiting the CYP2C9 enzyme, which suggested that piperine might be beneficial as an adjuvant to glimepiride in a proper dose, in diabetic patients. piperine 184-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 23156991-5 2012 The results revealed that a combination of glimepiride with piperine led to the enhancement of the bioavailability of glimepiride by inhibiting the CYP2C9 enzyme, which suggested that piperine might be beneficial as an adjuvant to glimepiride in a proper dose, in diabetic patients. glimepiride 118-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Chlorpropamide 296-310 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Chlorpropamide 296-310 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22886551-7 2012 These indicate a long-range haplotype containing the CYP2C9*13 and CYP2C19*2 mutation, which means most CYP2C9*13 carriers will carry the CYP2C19*2 allele and the six SNPs of the CYP2C9*1B haplotype group, and may have more reduced intrinsic clearance of drugs such as phenytoin, tolbutamide and chlorpropamide that are metabolized by both CYP2C9 and CYP2C19. Chlorpropamide 296-310 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 22735459-0 2012 Effects of CYP2C9*1/*3 and *1/*13 on the pharmacokinetics of losartan and its active metabolite E-3174. Losartan 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 22696420-7 2012 A direct comparison of SR-9186 and ketoconazole under typical assay conditions used in reaction phenotyping studies demonstrated that SR-9186 had selectivity over CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A5 greater than or equal to that of ketoconazole. 1-(4-imidazopyridinyl-7-phenyl)-3-(4'-cyanobiphenyl)urea 134-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 23331088-10 2012 Intriguingly, CYP2C19 may contributeto S-warfarin metabolism in patients, especially when CYP2C9 activity is compromised due to drug interactions orgenetic polymorphisms. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 22735459-6 2012 However, AUC0- of losartan was greater in CYP2C9*1/*3 subjects than in CYP2C9*1/*1, but these results were not significant (p < 0.05, but statistical power < 0.8). Losartan 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 22735459-6 2012 However, AUC0- of losartan was greater in CYP2C9*1/*3 subjects than in CYP2C9*1/*1, but these results were not significant (p < 0.05, but statistical power < 0.8). Losartan 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. losartan carboxylic acid 121-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. losartan carboxylic acid 121-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. losartan carboxylic acid 121-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. losartan carboxylic acid 121-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22735459-5 2012 The CYP2C9*1/*3 subjects showed lower oral clearance (p < 0.001) of losartan and higher Cmax (p < 0.01) and longer half-life (p < 0.01) of E-3174 than the CYP2C9*1/*1 subjects. Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 22735459-1 2012 OBJECTIVE: The effects of CYP2C9*1/*3 and *1/*13 genotypes were evaluated on the pharmacokinetics of losartan and its active metabolite, E-3174, in Korean subjects. Losartan 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. Losartan 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. Losartan 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22735459-4 2012 RESULTS: The CYP2C9*1/*13 subjects showed lower oral clearance (p < 0.001) and greater AUC0- (p < 0.01) of losartan and higher Cmax (p < 0.01) and longer half-life (p < 0.001) of E-3174 than the CYP2C9*1/*1 subjects, but AUC0- of E-3174 was not different. Losartan 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. Losartan 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22735459-9 2012 CONCLUSION: These results suggest that CYP2C9*1/*3 and CYP2C9*1/*13 are similarly associated with decreased formation of E-3174 from losartan, but the clinical effects of losartan may not be reduced by CYP2C9*1/*3 and CYP2C9*1/*13. Losartan 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 23167228-0 2012 The combined effects of clinical factors and CYP2C9 and VKORC1 gene polymorphisms on initiating warfarin treatment in patients after cardiac valve surgery. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 23167228-2 2012 The study aim was to analyze the influence of certain clinical factors and CYP2C9 and VKORC1 gene polymorphisms on the efficacy of initiation of warfarin treatment in patients after cardiac valve surgery. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22847201-0 2012 Lipid-lowering effect of fluvastatin in relation to cytochrome P450 2C9 variant alleles frequently distributed in the Czech population. Fluvastatin 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 22966884-3 2012 Results of a bivariate analysis demonstrated that among tested polymorphisms, carriers of the variant CYP2C9*3 tended to require significantly lower maintenance PHT dosages than wild-type carriers (p < 0.0001); on the other hand, carriers of the variants CYP2C9*3 or CYP2C19*3 revealed significantly higher CDRs than wild-type carriers (p < 0.004). Phenytoin 161-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 22966884-3 2012 Results of a bivariate analysis demonstrated that among tested polymorphisms, carriers of the variant CYP2C9*3 tended to require significantly lower maintenance PHT dosages than wild-type carriers (p < 0.0001); on the other hand, carriers of the variants CYP2C9*3 or CYP2C19*3 revealed significantly higher CDRs than wild-type carriers (p < 0.004). Phenytoin 161-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 258-264 22966884-3 2012 Results of a bivariate analysis demonstrated that among tested polymorphisms, carriers of the variant CYP2C9*3 tended to require significantly lower maintenance PHT dosages than wild-type carriers (p < 0.0001); on the other hand, carriers of the variants CYP2C9*3 or CYP2C19*3 revealed significantly higher CDRs than wild-type carriers (p < 0.004). cdrs 310-314 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 22966889-1 2012 Genotyping patients for CYP2C9 and VKORC1 polymorphisms can improve the accuracy of dosing during the initiation of anticoagulation with vitamin K antagonists (coumarin derivatives). Vitamin K 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 22966889-1 2012 Genotyping patients for CYP2C9 and VKORC1 polymorphisms can improve the accuracy of dosing during the initiation of anticoagulation with vitamin K antagonists (coumarin derivatives). coumarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 22538270-4 2012 The aim of the present study was to investigate the effect of gemfibrozil (CYP2C9 inhibitor) and its 1-O-beta-glucuronide (CYP2C8 inhibitor) on BRV disposition both in vivo (healthy participants) and in vitro (human liver microsomes and hepatocytes). Gemfibrozil 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22538270-11 2012 Overall, in vitro and in vivo data suggest that CYP2C8 and CYP2C9 are not involved in BRV hydroxylation, whereas hydroxylation of BRV-AC to BRV-OHAC is likely to be mediated by CYP2C9. brv-ac 130-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 22538270-11 2012 Overall, in vitro and in vivo data suggest that CYP2C8 and CYP2C9 are not involved in BRV hydroxylation, whereas hydroxylation of BRV-AC to BRV-OHAC is likely to be mediated by CYP2C9. brv-ohac 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 22743353-0 2012 Electrochemically driven drug metabolism via cytochrome P450 2C9 isozyme microsomes with cytochrome P450 reductase and indium tin oxide nanoparticle composites. indium tin oxide 119-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-64 22673786-5 2012 Ibuprofen"s clearance varies with CYP2C9*3 genotype. Ibuprofen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 22847201-1 2012 BACKGROUND: CYP2C9*3 allele has been reported to correlate with increased plasma concentration of fluvastatin active form in healthy volunteers. Fluvastatin 98-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 22847201-2 2012 We analyzed the correlation between the CYP2C9 genotype and cholesterol-lowering effect of fluvastatin in human hypercholesterolemic patients. Cholesterol 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 22847201-2 2012 We analyzed the correlation between the CYP2C9 genotype and cholesterol-lowering effect of fluvastatin in human hypercholesterolemic patients. Fluvastatin 91-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 22847201-4 2012 CYP2C9 genotype was determined by PCR-RFLP assay in 87 patients on concomitant fluvastatin therapy, in 48 patients on monotherapy, and in a control group of 254 healthy volunteers of Czech nationality. Fluvastatin 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 22847201-9 2012 The CYP2C9*1/*3 genotype was associated with a decrease in LDL-C levels (by 40.0% for CYP2C9*1/*3, but only by 22.4% for CYP2C9*1/*1), and with the reduction of TC (by 28.6% in CYP2C9*1/*3 versus 20.2% in CYP2C9*1/*1). Technetium 161-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 22847201-9 2012 The CYP2C9*1/*3 genotype was associated with a decrease in LDL-C levels (by 40.0% for CYP2C9*1/*3, but only by 22.4% for CYP2C9*1/*1), and with the reduction of TC (by 28.6% in CYP2C9*1/*3 versus 20.2% in CYP2C9*1/*1). Technetium 161-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22847201-9 2012 The CYP2C9*1/*3 genotype was associated with a decrease in LDL-C levels (by 40.0% for CYP2C9*1/*3, but only by 22.4% for CYP2C9*1/*1), and with the reduction of TC (by 28.6% in CYP2C9*1/*3 versus 20.2% in CYP2C9*1/*1). Technetium 161-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22847201-9 2012 The CYP2C9*1/*3 genotype was associated with a decrease in LDL-C levels (by 40.0% for CYP2C9*1/*3, but only by 22.4% for CYP2C9*1/*1), and with the reduction of TC (by 28.6% in CYP2C9*1/*3 versus 20.2% in CYP2C9*1/*1). Technetium 161-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22847201-9 2012 The CYP2C9*1/*3 genotype was associated with a decrease in LDL-C levels (by 40.0% for CYP2C9*1/*3, but only by 22.4% for CYP2C9*1/*1), and with the reduction of TC (by 28.6% in CYP2C9*1/*3 versus 20.2% in CYP2C9*1/*1). Technetium 161-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22847201-10 2012 CONCLUSIONS: In hypercholesterolemic patients, LDL-C serum concentration was decreased more significantly in fluvastatin-treated subjects bearing the CYP2C9*1/*3 genotype compared to CYP2C9*1/*1 genotype. Fluvastatin 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 22847201-10 2012 CONCLUSIONS: In hypercholesterolemic patients, LDL-C serum concentration was decreased more significantly in fluvastatin-treated subjects bearing the CYP2C9*1/*3 genotype compared to CYP2C9*1/*1 genotype. Fluvastatin 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22674970-2 2012 Polymorphisms in the cytochrome P450 (CYP) 2C9 and vitamin K epoxide reductase genes change warfarin pharmacokinetics and pharmacodynamics, respectively. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-46 21358752-1 2012 To optimize the warfarin dose, a population-specific pharmacogenomic algorithm was developed using multiple linear regression model with vitamin K intake and cytochrome P450 IIC polypeptide9 (CYP2C9(*)2 and (*)3), vitamin K epoxide reductase complex 1 (VKORC1(*)3, (*)4, D36Y and -1639 G>A) polymorphism profile of subjects who attained therapeutic international normalized ratio as predictors. Warfarin 16-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Flurbiprofen 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 21383771-4 2012 We confirmed the roles of three well-established loci (CYP2C9, VKORC1 and CYP4F2) in explaining warfarin dosage variation in the three Asian populations. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Flurbiprofen 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Flurbiprofen 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Tolbutamide 115-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Tolbutamide 115-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22547083-5 2012 METHODS: We assayed the activities of CYP2C9.1, CYP2C9.2, and CYP2C9.3 to metabolize diclofenac, flurbiprofen, and tolbutamide using a wild type or one of four POR variants (Q153R, A287P, R457H, and A503V). Tolbutamide 115-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22571356-2 2012 The effects of genetic polymorphisms at the vitamin K epoxide reductase [VKORC1] and cytochrome P450 2C9 [CYP2C9] loci have been increasingly acknowledged as contributory factors of enhanced warfarin sensitivity. Warfarin 191-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-104 22233204-8 2012 METHODS: In vitro experiments were conducted to assess the inhibition and induction potential of tofacitinib for major drug metabolizing enzymes (CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A4). tofacitinib 97-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 22360448-2 2012 CYP2C11 and CYP2C6 are considered as counterparts of human CYP2C9, which metabolizes many drugs including S-warfarin, diclofenac or ibuprofen. Warfarin 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 22360448-2 2012 CYP2C11 and CYP2C6 are considered as counterparts of human CYP2C9, which metabolizes many drugs including S-warfarin, diclofenac or ibuprofen. Diclofenac 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 22360448-2 2012 CYP2C11 and CYP2C6 are considered as counterparts of human CYP2C9, which metabolizes many drugs including S-warfarin, diclofenac or ibuprofen. Ibuprofen 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. Rosuvastatin Calcium 111-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. Rosuvastatin Calcium 172-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. Telmisartan 243-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. Sildenafil Citrate 256-266 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22360448-7 2012 Therefore, it should be taken into consideration that drugs metabolized by CYP2C9 in human could interact with rosuvastatin, as it has been already suggested for warfarin (rosuvastatin has increased its anticoagulant effect in human), and for telmisartan, sildenafil and glimepiride. glimepiride 271-282 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22571356-2 2012 The effects of genetic polymorphisms at the vitamin K epoxide reductase [VKORC1] and cytochrome P450 2C9 [CYP2C9] loci have been increasingly acknowledged as contributory factors of enhanced warfarin sensitivity. Warfarin 191-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 22496396-5 2012 CYP2C9 and CYP2C19 were the primary enzymes responsible for formation of the N-hydroxylated metabolite. Nitrogen 77-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23087510-0 2012 Effect of anti-tuberculosis therapy on polymorphic drug metabolizing enzyme CYP2C9 using phenytoin as a probe drug. Phenytoin 89-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 22531045-6 2012 In human liver microsomes, carnosic acid did not exhibit significant time-dependent inhibition for any of the cytochrome P450 enzymes investigated, although it did inhibit CYP2C9- and CYP3A4-catalyzed reactions with K(i) values of 9.2 and 4.3 muM, respectively. salvin 27-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 22294058-0 2012 Search for the molecular basis of ultra-rapid CYP2C9-catalysed metabolism: relationship between SNP IVS8-109A>T and the losartan metabolism phenotype in Swedes. Losartan 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 22288731-0 2012 Allele and genotype frequencies of CYP2C9 within an Iranian population (Mazandaran). mazandaran 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 23087510-7 2012 Phenotyping for CYP2C9 enzyme was done by measuring the ratio of phenytoin and its metabolite p-HPPH (para hydroxy phenyl hydantoin) by reverse phase HPLC (high performance liquid chromatography) method before and after one month of ATT. Phenytoin 65-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 23087510-7 2012 Phenotyping for CYP2C9 enzyme was done by measuring the ratio of phenytoin and its metabolite p-HPPH (para hydroxy phenyl hydantoin) by reverse phase HPLC (high performance liquid chromatography) method before and after one month of ATT. p-hpph 94-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 23087510-7 2012 Phenotyping for CYP2C9 enzyme was done by measuring the ratio of phenytoin and its metabolite p-HPPH (para hydroxy phenyl hydantoin) by reverse phase HPLC (high performance liquid chromatography) method before and after one month of ATT. para hydroxy phenyl hydantoin 102-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 23087510-8 2012 RESULTS: In the CYP2C9*1*1 genotype, the mean plasma concentrations of phenytoin before and after one month of ATT were 5.2 +- 0.3 mug/ml and 3.5 +- 0.4 mug/ml respectively, a reduction by 33% showing significant induction (P < 0.001). Phenytoin 71-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 22528326-5 2012 We also confirmed CYP2C9 *3 variant was related to lower warfarin dose (2.01 +- 0.23 mg/d) requirement compared to wild type (3.21 +- 0.11 mg/d) (P = 0.001). Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 22528326-7 2012 The multiple linear regression model including VKORC1-1639G>A, CYP2C9, CYP4F2 and clinical factors (body surface area (BSA) and age) could explain 42 % of the variance in the warfarin maintenance dose. Warfarin 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 21911247-1 2012 INTRODUCTION: The optimal dose of the oral anticoagulant warfarin varies with polymorphisms of the vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) genes. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-177 21911247-1 2012 INTRODUCTION: The optimal dose of the oral anticoagulant warfarin varies with polymorphisms of the vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) genes. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 22192158-2 2012 It is well known that gene polymorphisms of CytochromeP450 (CYP) 2C9 gene and the vitamin K epoxide reductase complex 1 (VKORC1) were significantly associated with warfarin dose. Warfarin 164-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-68 23157194-1 2012 Sulfonamide antimicrobials (sulfamethoxazole) contain an arylamine group, oxidized by CYP2C9 to the hydroxylamine with subsequent auto-oxidation to a highly reactive [-nitroso-] intermediate is a necessary (if not sufficient) cause of drug hypersensitivity. Sulfonamides 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22692316-6 2012 RESULTS: CYP1A2, CYP2C9, CYP2C19 and CYP3A4 in HLM were involved in bakuchiol metabolism, among which CYP2C19 showed the highest metabolic rate. bakuchiol 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 22692316-7 2012 Co-incubation with ABT (2.5 mmol/L) could inhibit more than 90% of the enzyme activities for CYP1A2, CYP2C9, CYP2C19 and CYP3A4. 1-aminobenzotriazole 19-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 23157194-1 2012 Sulfonamide antimicrobials (sulfamethoxazole) contain an arylamine group, oxidized by CYP2C9 to the hydroxylamine with subsequent auto-oxidation to a highly reactive [-nitroso-] intermediate is a necessary (if not sufficient) cause of drug hypersensitivity. Sulfamethoxazole 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22594566-1 2012 OBJECTIVE: Phenotyping cytochrome P450 (CYP) 2C9 activity with S-warfarin requires extensive blood sampling to characterize area under the concentration-time curve (AUC). Warfarin 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-48 22594566-2 2012 This retrospective data analysis was conducted to determine if truncated S-warfarin AUCs can be used to measure CYP2C9 activity. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 23157194-1 2012 Sulfonamide antimicrobials (sulfamethoxazole) contain an arylamine group, oxidized by CYP2C9 to the hydroxylamine with subsequent auto-oxidation to a highly reactive [-nitroso-] intermediate is a necessary (if not sufficient) cause of drug hypersensitivity. aniline 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 23157194-1 2012 Sulfonamide antimicrobials (sulfamethoxazole) contain an arylamine group, oxidized by CYP2C9 to the hydroxylamine with subsequent auto-oxidation to a highly reactive [-nitroso-] intermediate is a necessary (if not sufficient) cause of drug hypersensitivity. Hydroxylamine 100-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22561479-0 2012 The cytochrome P-450 2C9/2C19 but not the ABCB1 genetic polymorphism may be associated with the liver cytochrome 3A4 induction by phenytoin. Phenytoin 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-24 21811894-0 2012 Acenocoumarol sensitivity and pharmacokinetic characterization of CYP2C9 *5/*8,*8/*11,*9/*11 and VKORC1*2 in black African healthy Beninese subjects. Acenocoumarol 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 21811894-1 2012 This study aimed at investigating the contribution of CYP2C9 and VKORC1 genetic polymorphisms to inter-individual variability of acenocoumarol pharmacokinetics and pharmacodynamics in Black Africans from Benin. Acenocoumarol 129-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 21811894-8 2012 At 8 h post dose, the (S)-acenocoumarol concentrations in the CYP2C9*5/*8 and CYP2C9*9/*11 genotypes were about 1.9 and 5.1 fold higher compared with the CYP2C9*1/*1 genotype and 2.2- and 6.0-fold higher compared with the CYP2C9*1/*9 group, respectively. Acenocoumarol 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 21811894-8 2012 At 8 h post dose, the (S)-acenocoumarol concentrations in the CYP2C9*5/*8 and CYP2C9*9/*11 genotypes were about 1.9 and 5.1 fold higher compared with the CYP2C9*1/*1 genotype and 2.2- and 6.0-fold higher compared with the CYP2C9*1/*9 group, respectively. Acenocoumarol 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 21811894-8 2012 At 8 h post dose, the (S)-acenocoumarol concentrations in the CYP2C9*5/*8 and CYP2C9*9/*11 genotypes were about 1.9 and 5.1 fold higher compared with the CYP2C9*1/*1 genotype and 2.2- and 6.0-fold higher compared with the CYP2C9*1/*9 group, respectively. Acenocoumarol 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 21811894-8 2012 At 8 h post dose, the (S)-acenocoumarol concentrations in the CYP2C9*5/*8 and CYP2C9*9/*11 genotypes were about 1.9 and 5.1 fold higher compared with the CYP2C9*1/*1 genotype and 2.2- and 6.0-fold higher compared with the CYP2C9*1/*9 group, respectively. Acenocoumarol 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 21811894-11 2012 Significant association between plasma (S)-acenocoumarol concentration and CYP2C9 genotypes suggested the use of (S)-acenocoumarol for the phenotyping purpose. Acenocoumarol 39-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 21811894-11 2012 Significant association between plasma (S)-acenocoumarol concentration and CYP2C9 genotypes suggested the use of (S)-acenocoumarol for the phenotyping purpose. Acenocoumarol 113-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. vanillin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. vanillin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-118 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. ethyl vanillin 77-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 22450566-3 2012 The objective of the present work was to evaluate the impact of vanillin and ethyl vanillin on the activities of CYP2C9, CYP2E1, CYP3A4, CYP2B6 and CYP1A2 in human liver microsomes (HLM) in vitro, and impact on the activities of CYP1A2, CYP2C, CYP3A and CYP2E1 in rat liver microsomes (RLM) in vivo. ethyl vanillin 77-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-118 22328583-9 2012 Danazol inhibited CYP2C9, CYP2C8, and CYP2D6 with IC(50) values of 1.44, 1.95, and 2.74 muM, respectively. Danazol 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 22076061-1 2012 Molecular docking and molecular dynamics (MD) simulations are used to investigate the interactions of curcumin analogues (CAs) with human cytochrome P450 2 C9 (CYP2C9 or 2 C9) and the conformations of their binding sites. Curcumin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-158 22076061-1 2012 Molecular docking and molecular dynamics (MD) simulations are used to investigate the interactions of curcumin analogues (CAs) with human cytochrome P450 2 C9 (CYP2C9 or 2 C9) and the conformations of their binding sites. Curcumin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 22076061-3 2012 Results demonstrate that the most CAs studied lie 4~15 A above the heme of CYP2C9. cas 34-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22076061-3 2012 Results demonstrate that the most CAs studied lie 4~15 A above the heme of CYP2C9. Heme 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 22076061-4 2012 The presence of CAs makes some residues in bound CYP2C9s become more flexible. cas 16-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 22311023-1 2012 Metabolic transformations of two substrates for human cytochrome P450 (CYP450) 2C9, tolbutamide and diclofenac, were investigated in hepatic microsomes from Atlantic salmon (Salmo salar L.). Tolbutamide 84-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-82 22311023-1 2012 Metabolic transformations of two substrates for human cytochrome P450 (CYP450) 2C9, tolbutamide and diclofenac, were investigated in hepatic microsomes from Atlantic salmon (Salmo salar L.). Diclofenac 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-82 22311023-4 2012 Addition of sulfaphenazole, an inhibitor for mammalian CYP2C9, in a range from 1 to 200 muM decreased formation of TBOH in a concentration-dependent manner, but not to 50%. Sulfaphenazole 12-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22311023-4 2012 Addition of sulfaphenazole, an inhibitor for mammalian CYP2C9, in a range from 1 to 200 muM decreased formation of TBOH in a concentration-dependent manner, but not to 50%. tboh 115-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22534826-0 2012 A new algorithm to predict warfarin dose from polymorphisms of CYP4F2 , CYP2C9 and VKORC1 and clinical variables: derivation in Han Chinese patients with non valvular atrial fibrillation. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 22534826-2 2012 The objective of this study was to develop a new algorithm by polymorphisms of CYP2C9, VKORC1 and CYP4F2 to predict the daily stable dose of warfarin in Chinese patients with NVAF. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 22549502-7 2012 RESULTS: From multiple linear regression models, vitamin K epoxide reductase complex subunit 1, CYP2C9, CYP4F2, and age were found to have significant effects on warfarin stable dose. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 22436668-0 2012 Simultaneous determination of fluoxetine and norfluoxetine enantiomers using isotope discrimination mass spectroscopy solution method and its application in the CYP2C9-mediated stereoselective interactions. Fluoxetine 30-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 22436668-0 2012 Simultaneous determination of fluoxetine and norfluoxetine enantiomers using isotope discrimination mass spectroscopy solution method and its application in the CYP2C9-mediated stereoselective interactions. norfluoxetine 45-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 22436668-1 2012 In this study, we developed an LC-MS/MS method based on an isotope discrimination mass spectroscopy solution (IDMSS) technology to simultaneously quantify enantiomers of fluoxetine (FLX) and norfluoxetine (NFLX) in a CYP2C9 incubation mixture. Fluoxetine 170-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 22436668-1 2012 In this study, we developed an LC-MS/MS method based on an isotope discrimination mass spectroscopy solution (IDMSS) technology to simultaneously quantify enantiomers of fluoxetine (FLX) and norfluoxetine (NFLX) in a CYP2C9 incubation mixture. norfluoxetine 191-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 22436668-8 2012 No significant kinetic isotope effect was found in the metabolism of S-FLX-d5 catalyzed by CYP2C9*1 and CYP2C9*2. s-flx-d5 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 22436668-9 2012 The half-maximal inhibitory concentration values between R-FLX and S-FLX catalyzed by CYP2C9*1 and CYP2C9*2 were determined in this study. r-flx 57-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22436668-9 2012 The half-maximal inhibitory concentration values between R-FLX and S-FLX catalyzed by CYP2C9*1 and CYP2C9*2 were determined in this study. r-flx 57-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22436668-9 2012 The half-maximal inhibitory concentration values between R-FLX and S-FLX catalyzed by CYP2C9*1 and CYP2C9*2 were determined in this study. Fluoxetine 67-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22436668-9 2012 The half-maximal inhibitory concentration values between R-FLX and S-FLX catalyzed by CYP2C9*1 and CYP2C9*2 were determined in this study. Fluoxetine 67-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. r- to s-flx 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. r- to s-flx 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. Sulfur 32-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. Sulfur 32-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. r-flx 72-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22436668-10 2012 The inhibitory effects of R- to S-FLX were stronger than those of S- to R-FLX in both CYP2C9*1 and CYP2C9*2. r-flx 72-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22943641-2 2012 METHODS: CYP2C9*3 (1075A/C, rs1057910) and VKORC1 (-1639A/G, rs9923231) genotyping was detected in 100 patients receiving Warfarin therapy by the newly developed HRM method and traditional genotyping methods including PCR-restriction fragment length polymorphism (PCR-RFLP), TaqMan probe and DNA sequencing. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 22331994-1 2012 Long-chain unsaturated fatty acids inhibit several cytochrome P450 and UDP-glucuronosyltransferase (UGT) enzymes involved in drug metabolism, including CYP2C8, CYP2C9, UGT1A9, UGT2B4, and UGT2B7. long-chain unsaturated fatty acids 0-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 22071881-0 2012 Factor VII R353Q genetic polymorphism is associated with altered warfarin sensitivity among CYP2C9 *1/*1 carriers. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 22071881-13 2012 The prospective use of this polymorphism, along with CYP2C9 and VKORC1, may enhance the accuracy of warfarin loading. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 22483397-2 2012 CYP isoform specific substrates and their metabolites of CYP1A2 (caffeine), CYP2C9 (losartan), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan) and CYP3A (midazolam) were all simultaneously analyzed using LC-MS/MS after administration of a mixture of five drugs (i.e., a "cocktail approach") to healthy volunteers. Losartan 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 22108774-1 2012 PURPOSE: Zafirlukast is a substrate of cytochrome P450 2C9 (CYP2C9) and cytochrome P450 3A4 (CYP3A4) in vitro, but the role of these enzymes in its metabolism in vivo is unknown. zafirlukast 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 22108774-1 2012 PURPOSE: Zafirlukast is a substrate of cytochrome P450 2C9 (CYP2C9) and cytochrome P450 3A4 (CYP3A4) in vitro, but the role of these enzymes in its metabolism in vivo is unknown. zafirlukast 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 22108774-7 2012 CONCLUSIONS: Fluconazole, but not itraconazole, increases zafirlukast plasma concentrations, strongly suggesting that CYP2C9 but not CYP3A4 participates in zafirlukast metabolism in humans. Fluconazole 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 22108774-7 2012 CONCLUSIONS: Fluconazole, but not itraconazole, increases zafirlukast plasma concentrations, strongly suggesting that CYP2C9 but not CYP3A4 participates in zafirlukast metabolism in humans. zafirlukast 156-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 22409277-0 2012 An evaluation of gene-gene interaction between the CYP2C9 and VKORC1 genotypes affecting the anticoagulant effect of phenprocoumon and acenocoumarol. Phenprocoumon 117-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 22409277-0 2012 An evaluation of gene-gene interaction between the CYP2C9 and VKORC1 genotypes affecting the anticoagulant effect of phenprocoumon and acenocoumarol. Acenocoumarol 135-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 22409277-2 2012 OBJECTIVES: We aimed to provide a definite answer regarding the question whether there exists a gene-gene interaction between the CYP2C9 and VKORC1 genotypes affecting the anticoagulant effect of phenprocoumon and acenocoumarol. Phenprocoumon 196-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 22409277-2 2012 OBJECTIVES: We aimed to provide a definite answer regarding the question whether there exists a gene-gene interaction between the CYP2C9 and VKORC1 genotypes affecting the anticoagulant effect of phenprocoumon and acenocoumarol. Acenocoumarol 214-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 21814747-8 2012 Norendoxifen inhibited placental aromatase with an IC(50) of 90 nM, while it inhibited human liver CYP2C9 and CYP3A with IC(50) values of 990 and 908 nM, respectively. N,N-didesmethyl-4-hydroxytamoxifen 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22594507-0 2012 A clinically significant interaction between warfarin and simvastatin is unique to carriers of the CYP2C9*3 allele. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22594507-0 2012 A clinically significant interaction between warfarin and simvastatin is unique to carriers of the CYP2C9*3 allele. Simvastatin 58-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22594507-2 2012 PATIENTS & METHODS: The influence of the CYP2C9*2 and CYP2C9*3 polymorphisms on the interaction between simvastatin and warfarin was analyzed in data from 1132 patients. Simvastatin 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 22594507-2 2012 PATIENTS & METHODS: The influence of the CYP2C9*2 and CYP2C9*3 polymorphisms on the interaction between simvastatin and warfarin was analyzed in data from 1132 patients. Simvastatin 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 22594507-3 2012 RESULTS: Simvastatin use reduced warfarin dose requirements by 29% in carriers of the CYP2C9*3 allele, compared with 5% in noncarriers. Simvastatin 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22594507-3 2012 RESULTS: Simvastatin use reduced warfarin dose requirements by 29% in carriers of the CYP2C9*3 allele, compared with 5% in noncarriers. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 22594507-4 2012 A regression model showed a significant influence of CYP2C9*3 on the drug-drug interaction, predicting a warfarin dose reduction of 25% in CYP2C9*3 heterozygotes and 43% in CYP2C9*3 homozygotes. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 22594507-4 2012 A regression model showed a significant influence of CYP2C9*3 on the drug-drug interaction, predicting a warfarin dose reduction of 25% in CYP2C9*3 heterozygotes and 43% in CYP2C9*3 homozygotes. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 22594507-4 2012 A regression model showed a significant influence of CYP2C9*3 on the drug-drug interaction, predicting a warfarin dose reduction of 25% in CYP2C9*3 heterozygotes and 43% in CYP2C9*3 homozygotes. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 22594507-5 2012 CONCLUSION: Our data indicate that the CYP2C9*3 polymorphism predisposes for a pharmacologic interaction between warfarin and simvastatin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 22594507-5 2012 CONCLUSION: Our data indicate that the CYP2C9*3 polymorphism predisposes for a pharmacologic interaction between warfarin and simvastatin. Simvastatin 126-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 22266406-3 2012 RESULTS: We confirm the association between high-frequency SNPs, VKORC1 c.-1639G>A and CYP2C9 *2/*3 and warfarin sensitivity, and contribute additional evidence that the VKORC1 p.Asp36Tyr variant is recurrent and independently associated with warfarin resistance in our population. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 22378156-0 2012 Decreased warfarin clearance associated with the CYP2C9 R150H (*8) polymorphism. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 22378156-1 2012 The cytochrome P450 (CYP) 2C9 R150H (*8) allele occurs commonly in African Americans and is associated with lower warfarin dose requirements. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-29 22378156-2 2012 We conducted a pharmacokinetic study to examine whether the CYP2C9*8 allele impacts warfarin clearance in African-American patients. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 22378156-4 2012 We observed a 30% reduction in the unbound oral clearance of S-warfarin and a 25% lower R- to S-warfarin plasma concentration ratio in patients with the CYP2C9*8 allele (n = 12) as compared to CYP2C9*1 homozygotes (n = 26). Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 22378156-4 2012 We observed a 30% reduction in the unbound oral clearance of S-warfarin and a 25% lower R- to S-warfarin plasma concentration ratio in patients with the CYP2C9*8 allele (n = 12) as compared to CYP2C9*1 homozygotes (n = 26). Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 22378156-6 2012 These data show that the R150H variant protein expressed by the CYP2C9*8 allele is associated with lower S-warfarin clearance. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 22378156-7 2012 This finding provides clinical and experimental evidence to explain the lower warfarin dose requirements in patients with the CYP2C9*8 allele. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 22205778-4 2012 To this end, the selective CYP2C9 mechanism-based inactivator tienilic acid was used to reduce irreversibly the total CYP2C9 activity in human liver microsomes. Ticrynafen 62-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 22205778-4 2012 To this end, the selective CYP2C9 mechanism-based inactivator tienilic acid was used to reduce irreversibly the total CYP2C9 activity in human liver microsomes. Ticrynafen 62-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 22205778-5 2012 Tienilic acid concentrations were effectively titrated to produce microsomal preparations with 43 and 73% less activity, mimicking the CYP2C9*1/*3 and CYP2C9*3/*3 genotypes, respectively. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 22205778-5 2012 Tienilic acid concentrations were effectively titrated to produce microsomal preparations with 43 and 73% less activity, mimicking the CYP2C9*1/*3 and CYP2C9*3/*3 genotypes, respectively. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 22205778-7 2012 In lieu of using rare human liver microsomes from CYP2C9*1/*3 and CYP2C9*3/*3 individuals, a tienilic acid-created knockdown in human liver microsomes may be an appropriate in vitro model to determine CYP2C9-mediated metabolism of a given substrate, to determine whether other drug-metabolizing enzymes may compensate for reduced CYP2C9 activity, and to predict the extent of genotype-dependent drug-drug interactions. Ticrynafen 93-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 22205778-7 2012 In lieu of using rare human liver microsomes from CYP2C9*1/*3 and CYP2C9*3/*3 individuals, a tienilic acid-created knockdown in human liver microsomes may be an appropriate in vitro model to determine CYP2C9-mediated metabolism of a given substrate, to determine whether other drug-metabolizing enzymes may compensate for reduced CYP2C9 activity, and to predict the extent of genotype-dependent drug-drug interactions. Ticrynafen 93-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 22205778-7 2012 In lieu of using rare human liver microsomes from CYP2C9*1/*3 and CYP2C9*3/*3 individuals, a tienilic acid-created knockdown in human liver microsomes may be an appropriate in vitro model to determine CYP2C9-mediated metabolism of a given substrate, to determine whether other drug-metabolizing enzymes may compensate for reduced CYP2C9 activity, and to predict the extent of genotype-dependent drug-drug interactions. Ticrynafen 93-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 22329513-0 2012 Differential oxidation of two thiophene-containing regioisomers to reactive metabolites by cytochrome P450 2C9. Thiophenes 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-110 22317799-4 2012 Ideally, such an assay would use a single PCR reaction and, without further processing, a single microchip electrophoresis (ME) run to determine the 3 single-nucleotide polymorphisms (SNPs) affecting warfarin sensitivity [i.e., CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) *2, CYP2C9 *3, and the VKORC1 (vitamin K epoxide reductase complex 1) A/B haplotype]. Warfarin 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 228-234 22317799-4 2012 Ideally, such an assay would use a single PCR reaction and, without further processing, a single microchip electrophoresis (ME) run to determine the 3 single-nucleotide polymorphisms (SNPs) affecting warfarin sensitivity [i.e., CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) *2, CYP2C9 *3, and the VKORC1 (vitamin K epoxide reductase complex 1) A/B haplotype]. Warfarin 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 22266406-3 2012 RESULTS: We confirm the association between high-frequency SNPs, VKORC1 c.-1639G>A and CYP2C9 *2/*3 and warfarin sensitivity, and contribute additional evidence that the VKORC1 p.Asp36Tyr variant is recurrent and independently associated with warfarin resistance in our population. Warfarin 246-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 22266406-5 2012 CONCLUSION: Warfarin sensitivity was predicted by known VKORC1 and CYP2C9 SNPs. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 22269145-6 2012 Two monohydroxylated metabolites of lower abundance were formed by CYP3A4, and interestingly, although CYP2C9 showed no activity toward the parent compound, this enzyme appeared to act in concert with CYP3A4 to form two minor dihydroxylated products of ETR. etravirine 253-256 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 22589111-0 2012 Rapid testing of clopidogrel resistance by genotyping of CYP2C19 and CYP2C9 polymorphisms using denaturing on-chip capillary electrophoresis. Clopidogrel 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 22269145-10 2012 Taken together, these data suggest that ETR and ETR metabolites are substrates of CYP2C19, CYP3A4, CYP2C9, UGT1A3, and UGT1A8 and that ETR is a PXR-dependent modulator of CYP3A4 mRNA levels. etravirine 40-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22269145-10 2012 Taken together, these data suggest that ETR and ETR metabolites are substrates of CYP2C19, CYP3A4, CYP2C9, UGT1A3, and UGT1A8 and that ETR is a PXR-dependent modulator of CYP3A4 mRNA levels. etravirine 48-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22269145-10 2012 Taken together, these data suggest that ETR and ETR metabolites are substrates of CYP2C19, CYP3A4, CYP2C9, UGT1A3, and UGT1A8 and that ETR is a PXR-dependent modulator of CYP3A4 mRNA levels. etravirine 48-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 22589111-4 2012 Genotyping of CYP2C19 and CYP2C9 polymorphisms allows to identify slow metabolizers showing resistance to clopidogrel therapy. Clopidogrel 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 22334041-10 2012 In single-SNP adjusted analysis, nine SNPs in the XPC, CYP2C9, PAX4, MTRR, and GAN genes were associated with cyclosporine nephrotoxicity. Cyclosporine 110-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 22270321-0 2012 Population pharmacokinetic modelling of S-warfarin to evaluate the design of drug-drug interaction studies for CYP2C9. Sulfur 40-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 22270321-1 2012 This study is to assess pharmacokinetic (PK) sampling time schedules and trial size requirements of drug-drug interaction (DDI) studies for CYP2C9, based on S-warfarin population PK models. Warfarin 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 21507031-0 2012 Clinical relevance of VKORC1 (G-1639A and C1173T) and CYP2C9*3 among patients on warfarin. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 21507031-1 2012 WHAT IS KNOWN AND OBJECTIVES: Testing for cytochrome P450-2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) variant alleles is recommended by the FDA for dosing of warfarin. Warfarin 186-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-61 21507031-1 2012 WHAT IS KNOWN AND OBJECTIVES: Testing for cytochrome P450-2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) variant alleles is recommended by the FDA for dosing of warfarin. Warfarin 186-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21507031-3 2012 We therefore studied the impact of genetic polymorphisms of CYP2C9 and VKORC1 on warfarin dose requirement in Malaysia. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 21507031-8 2012 RESULTS AND DISCUSSION: A model which included age and variants of CYP2C9 and VKORC1 account for about 37% of the variability in warfarin dose required to achieve INR of 2-4. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 22252093-0 2012 Long-term anticoagulant effects of the CYP2C9 and VKORC1 genotypes in acenocoumarol users. Acenocoumarol 70-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 22252093-1 2012 BACKGROUND: The required acenocoumarol dose and the risk of underanticoagulation and overanticoagulation are associated with the CYP2C9 and VKORC1 genotypes. Acenocoumarol 25-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 22252093-5 2012 RESULTS: Patients with polymorphisms in CYP2C9 and VKORC1 had a higher risk of overanticoagulation (up to 74%) and a lower risk of underanticoagulation (down to 45%) in the first month of treatment with acenocoumarol, but this effect diminished after 1-6 months. Acenocoumarol 203-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 22459452-3 2012 Hence there are chances of drug-drug interaction because modulations of isoenzymes involved in metabolism of phenytoin and aceclofenac are CYP2C9/10 and CYP2C19. Phenytoin 109-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 22459452-3 2012 Hence there are chances of drug-drug interaction because modulations of isoenzymes involved in metabolism of phenytoin and aceclofenac are CYP2C9/10 and CYP2C19. aceclofenac 123-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 22321050-1 2012 We recently designed the CIME cocktail consisting of 10 drugs to assess the activity of the major human CYPs (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP3A), a phase II enzyme (UGT1A1/6/9), two drug transporters (P-gp and OATP1B1) and a component of the renal function ( Videau et al. cime 25-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 22364953-8 2012 An overrepresentation was seen for poly-aromatic hydrocarbons (group of procarcinogens) among CYP2C9 active and mutagenic compounds compared to CYP2C9 inactive and mutagenic compounds. poly-aromatic hydrocarbons 35-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 21881887-6 2012 INR levels and S-warfarin concentrations were associated with CYP2C9 and VKORC1 genotypes. Warfarin 17-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 21883387-0 2012 Impact of genetic factors (VKORC1, CYP2C9, CYP4F2 and EPHX1) on the anticoagulation response to fluindione. fluindione 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 21883387-1 2012 AIM: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9) and of a key pharmacologic target of vitamin K antagonists, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to coumarin derivatives. Vitamin K 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-83 21883387-1 2012 AIM: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9) and of a key pharmacologic target of vitamin K antagonists, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to coumarin derivatives. Vitamin K 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21883387-1 2012 AIM: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9) and of a key pharmacologic target of vitamin K antagonists, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to coumarin derivatives. coumarin 243-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-83 21883387-1 2012 AIM: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9) and of a key pharmacologic target of vitamin K antagonists, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to coumarin derivatives. coumarin 243-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21820929-6 2012 RESULTS: CYP3A4 was responsible for the metabolism of lovastatin, simvastatin and atorvastatin; fluvastatin depends on CYP2C9; P-glycoprotein is responsible for decreased atorvastatin, pravastatin, simvastatin and lovastatin concentrations. Fluvastatin 96-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 22198820-5 2012 RESULTS: Single-nucleotide polymorphism association analysis showed that VKORC1, CYP2C9 and CYP4F2 variations were highly associated with the warfarin maintenance dose. Warfarin 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 21935704-5 2012 METHODS: In a cross-sectional study, we interviewed 244 patients in the maintenance phase of warfarin therapy and detected polymorphisms in the VKORC1 and CYP2C9 genes. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 22198820-7 2012 The final pharmacogenetic equation explained 43.65% of interindividual variation of warfarin maintenance dose with age, body surface area, VKORC1 g.3588G>A, CYP2C9*3, CYP4F2 c.1297G>A, amiodarone, fluconazole, and diltiazem accounting for 1.97, 2.74, 24.12, 3.94, 1.64, 5.92, 2.47, and 0.84% of variation. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 22198820-8 2012 CONCLUSION: The present study indicated that VKORC1, CYP4F2, and CYP2C9 genotypes and interacting drugs had a significant impact on the warfarin maintenance dose in Chinese patients with heart valve replacement and demonstrated that integrating interacting drugs can largely improve the predictability of the dose algorithm. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 22022918-6 2012 This reaction was inhibited significantly by ketoconazole, a CYP3A inhibitor, and azamulin, a mechanism-based inhibitor of CYP3A, while no substantial effect was observed using selective inhibitors for eight other P450s (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1). Ketoconazole 45-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 253-259 22186153-5 2012 Human CYP2C9(*)1, CYP2C19 and rat CYP2C6 were the major CYP isoforms producing 5-OH MEHP and 5-Oxo MEHP metabolites; however, only human CYP2C9(*)1 and 2C9(*)2 produced 5-carboxy MEPP from MEHP. 5-oh mehp 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 22186153-5 2012 Human CYP2C9(*)1, CYP2C19 and rat CYP2C6 were the major CYP isoforms producing 5-OH MEHP and 5-Oxo MEHP metabolites; however, only human CYP2C9(*)1 and 2C9(*)2 produced 5-carboxy MEPP from MEHP. 5-oxo mehp 93-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 22186153-5 2012 Human CYP2C9(*)1, CYP2C19 and rat CYP2C6 were the major CYP isoforms producing 5-OH MEHP and 5-Oxo MEHP metabolites; however, only human CYP2C9(*)1 and 2C9(*)2 produced 5-carboxy MEPP from MEHP. 5-carboxy mepp 169-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 22186153-5 2012 Human CYP2C9(*)1, CYP2C19 and rat CYP2C6 were the major CYP isoforms producing 5-OH MEHP and 5-Oxo MEHP metabolites; however, only human CYP2C9(*)1 and 2C9(*)2 produced 5-carboxy MEPP from MEHP. mono-(2-ethylhexyl)phthalate 84-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 22186153-7 2012 Percent total normalized rates (%TNR) by CYP2C9(*)1 in human liver microsomes (HLM) were 94%, 98% and 100%, respectively, for 5-OH MEHP, 5-Oxo MEHP, 5-carboxy MEPP, and 76% for PA production by CYP3A4. 5-oh mehp 126-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 22186153-7 2012 Percent total normalized rates (%TNR) by CYP2C9(*)1 in human liver microsomes (HLM) were 94%, 98% and 100%, respectively, for 5-OH MEHP, 5-Oxo MEHP, 5-carboxy MEPP, and 76% for PA production by CYP3A4. 5-carboxy mepp 149-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 22379999-2 2012 Warfarin is a typical example of where pharmacogenetics could help the individual patient by modeling the dose, based on clinical factors and genetic variation in CYP2C9 and VKORC1. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 22022918-6 2012 This reaction was inhibited significantly by ketoconazole, a CYP3A inhibitor, and azamulin, a mechanism-based inhibitor of CYP3A, while no substantial effect was observed using selective inhibitors for eight other P450s (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1). azamulin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 253-259 22117526-5 2012 Comparing two assessment results for CYP2D6 and CYP2C9 inhibitors, the FDA assessment suggested potential DDI risks for sulphinpyrazone and amitriptyline, while the EMEA assessment indicated no potential risk for these drugs. Sulfinpyrazone 120-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22226725-2 2012 The only human CYP known to produce relevant amounts of 3-OH-Ibu is CYP2C9 and as genetic polymorphisms of CYP2C9 influence the metabolization of numerous drugs, the availability of reference standards for CYP2C9-specific metabolites is of considerable interest. 3-oh-ibu 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 22226725-2 2012 The only human CYP known to produce relevant amounts of 3-OH-Ibu is CYP2C9 and as genetic polymorphisms of CYP2C9 influence the metabolization of numerous drugs, the availability of reference standards for CYP2C9-specific metabolites is of considerable interest. 3-oh-ibu 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 22226725-2 2012 The only human CYP known to produce relevant amounts of 3-OH-Ibu is CYP2C9 and as genetic polymorphisms of CYP2C9 influence the metabolization of numerous drugs, the availability of reference standards for CYP2C9-specific metabolites is of considerable interest. 3-oh-ibu 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 22226725-4 2012 The recombinant fission yeast strain CAD68 coexpressing human CYP2C9 and CPR was used for the whole-cell biotransformation of Ibu to 3-OH-Ibu in 1L batch-scale for 75h. Ibuprofen 126-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 22226725-4 2012 The recombinant fission yeast strain CAD68 coexpressing human CYP2C9 and CPR was used for the whole-cell biotransformation of Ibu to 3-OH-Ibu in 1L batch-scale for 75h. 3-oh-ibu 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 22274142-0 2012 Prediction of warfarin dose reductions in Puerto Rican patients, based on combinatorial CYP2C9 and VKORC1 genotypes. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 22274142-1 2012 BACKGROUND: The influence of CYP2C9 and VKORC1 polymorphisms on warfarin dose has been investigated in white, Asian, and African American populations but not in Puerto Rican Hispanic patients. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 22274142-10 2012 Among patients with a given CYP2C9 genotype, warfarin dose requirements declined from GG to AA haplotypes; whereas, within each VKORC1 haplotype, the dose decreased as the number of CYP2C9 variants increased. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 21838784-1 2012 AIM: According to product information, montelukast is extensively metabolized by CYP3A4 and CYP2C9. montelukast 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 21842338-0 2012 CYP2C9 3 and 13 alleles significantly affect the pharmacokinetics of irbesartan in healthy Korean subjects. Irbesartan 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21842338-1 2012 PURPOSE: To evaluate the effects of two major polymorphisms of CYP2C9, CYP2C9 3 and CYP2C9 13, on the pharmacokinetics of irbesartan in healthy Korean volunteers. Irbesartan 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21842338-1 2012 PURPOSE: To evaluate the effects of two major polymorphisms of CYP2C9, CYP2C9 3 and CYP2C9 13, on the pharmacokinetics of irbesartan in healthy Korean volunteers. Irbesartan 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 21842338-1 2012 PURPOSE: To evaluate the effects of two major polymorphisms of CYP2C9, CYP2C9 3 and CYP2C9 13, on the pharmacokinetics of irbesartan in healthy Korean volunteers. Irbesartan 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 21842338-8 2012 CONCLUSIONS: CYP2C9 genetic polymorphisms markedly affected the pharmacokinetics of irbesartan in this study sample. Irbesartan 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 21842338-9 2012 The CYP2C9 3 and CYP2C9 13 alleles appear to be associated with the decreased metabolism of irbesartan. Irbesartan 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21842338-9 2012 The CYP2C9 3 and CYP2C9 13 alleles appear to be associated with the decreased metabolism of irbesartan. Irbesartan 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 21870106-7 2012 In addition, losartan/E-3174 ratio doubled (P < 0.01) after BBR administration, indicating a decrease in CYP2C9 activity. Losartan 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 21870106-7 2012 In addition, losartan/E-3174 ratio doubled (P < 0.01) after BBR administration, indicating a decrease in CYP2C9 activity. losartan carboxylic acid 22-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 21208246-0 2012 Frequency of CYP2C9 variant alleles, including CYP2C9*13 in a Korean population and effect on glimepiride pharmacokinetics. glimepiride 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 21208246-11 2012 Our data on the one subject with this genotype suggest that CYP2C9*3/*3 momozygotes have lower clearance of glimepiride and are exposed to higher levels of the drug than wild-type homozygotes. glimepiride 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 22188360-6 2012 CONCLUSION: These data indicate that copy number variants in the principal genes involved in warfarin dose variability (CYP2C9, VKORC1), including genes with lesser effect (CYP4F2, GGCX), and those that may be more relevant among certain racial groups (CALU), are rare in multiethnic populations, including African-Americans. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 22106207-2 2012 The objective of this study was to examine the impact of N-acetyltransferase 2 (NAT2) and CYP2C9 polymorphisms on the pharmacokinetics of SMX in 118 renal transplant recipients. Sulfamethoxazole 138-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 22452429-0 2012 Warfarin pharmacogenetics: polymorphisms of the CYP2C9, CYP4F2, and VKORC1 loci in a genetically admixed Omani population. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22248286-7 2012 CONCLUSION: We developed a pharmacogenetic dose algorithm for warfarin treatment that uses genotypes from two genes (VKORC1 and CYP2C9) and clinical variables to predict therapeutic maintenance doses in Chinese patients with venous thromboembolism. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 22117526-5 2012 Comparing two assessment results for CYP2D6 and CYP2C9 inhibitors, the FDA assessment suggested potential DDI risks for sulphinpyrazone and amitriptyline, while the EMEA assessment indicated no potential risk for these drugs. Amitriptyline 140-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22149257-0 2012 A pharmacokinetic-pharmacodynamic model for predicting the impact of CYP2C9 and VKORC1 polymorphisms on fluindione and acenocoumarol during induction therapy. fluindione 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 22130800-4 2012 The results showed that height, target international normalized ratio and VKORC1 and CYP2C9 genotypes were the main determinants of warfarin dose requirement, accounting for 48.1%, 4.4%, 18.2%, and 2.0% of variability, respectively, and explaining 69.7% of the variability. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21692828-10 2012 RESULTS: Bodyweight, age, sex and CYP2C9 genotype significantly influenced S-warfarin clearance. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 22149257-12 2012 CONCLUSION: During initiation of therapy, CYP2C9 and VKORC1 genetic polymorphisms are important predictors of fluindione and acenocoumarol pharmacokinetic-pharmacodynamic responses. fluindione 110-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 22010099-0 2012 VKORC1 and CYP2C9 genotype and patient characteristics explain a large proportion of the variability in warfarin dose requirement among children. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 22010099-4 2012 Multiple regression analysis of the data showed that, although CYP4F2 made no contribution to the dose model, 72.4% of the variability in warfarin dose requirement is attributed to by patient height, genetic polymorphisms in VKORC1 and CYP2C9, and indication for warfarin. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 236-242 22293514-3 2012 Etravirine, a second-generation non-nucleoside reverse transcriptase inhibitor has been shown to induce CYP3A4, while inhibiting CYP2C9 and CYP2C19. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 22149257-12 2012 CONCLUSION: During initiation of therapy, CYP2C9 and VKORC1 genetic polymorphisms are important predictors of fluindione and acenocoumarol pharmacokinetic-pharmacodynamic responses. Acenocoumarol 125-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 22149257-0 2012 A pharmacokinetic-pharmacodynamic model for predicting the impact of CYP2C9 and VKORC1 polymorphisms on fluindione and acenocoumarol during induction therapy. Acenocoumarol 119-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 22447115-4 2012 When cells were treated with a typical CYP3A substrate (triazolam), CYP2C9 substrate (diclofenac) or UGT1A1 substrate (SN-38), large amounts of their metabolites were detected in the medium of cells cultured on micro-space cell culture plates. Diclofenac 86-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 21890570-9 2012 It is hypothesized that warfarin and dapsone compete for binding on the CYP2C9 and CYP3A4 isoenzymes and therefore serum concentration of warfarin was elevated. Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21890570-9 2012 It is hypothesized that warfarin and dapsone compete for binding on the CYP2C9 and CYP3A4 isoenzymes and therefore serum concentration of warfarin was elevated. Dapsone 37-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21890570-9 2012 It is hypothesized that warfarin and dapsone compete for binding on the CYP2C9 and CYP3A4 isoenzymes and therefore serum concentration of warfarin was elevated. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Triazolam 34-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Diclofenac 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Irinotecan 60-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 23093260-0 2012 Development of a HPLC method for the determination of losartan urinary metabolic ratio to be used for the determination of CYP2C9 hydroxylation phenotypes. Losartan 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 22447115-5 2012 The formation of metabolites from triazolam, diclofenac and SN-38 was strongly inhibited by co-treatment with a CYP3A inhibitor (ketoconazole), CYP2C9 inhibitor (sulfaphenazole) and UGT1A1 inhibitor (ketoconazole), respectively. Sulfaphenazole 162-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 23093260-1 2012 BACKGROUND: Losartan is metabolized to losartan carboxylic acid (E-3174) by the polymorphic cytochrome CYP2C9. Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 23093260-1 2012 BACKGROUND: Losartan is metabolized to losartan carboxylic acid (E-3174) by the polymorphic cytochrome CYP2C9. losartan carboxylic acid 39-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 22375877-2 2012 In this study, we examined the inhibitory potentials of capsaicin and dihydrocapsaicin against CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 activities in human liver microsomes. Capsaicin 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 23093260-1 2012 BACKGROUND: Losartan is metabolized to losartan carboxylic acid (E-3174) by the polymorphic cytochrome CYP2C9. losartan carboxylic acid 65-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 23093260-6 2012 Analyses with the present HPLC method show significant differences (p<0.05) in losartan MRs between the four CYP2C9 genotype groups in 13 Spanish healthy volunteers. Losartan 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 23093260-8 2012 Therefore, it appears to be useful for CYP2C9 phenotyping using losartan as a drug test in populations, such as Hispanics with different allele combinations. Losartan 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 23099353-0 2012 Effects of UGT1A6, UGT2B7, and CYP2C9 genotypes on plasma concentrations of valproic acid in Chinese children with epilepsy. Valproic Acid 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 22375877-2 2012 In this study, we examined the inhibitory potentials of capsaicin and dihydrocapsaicin against CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 activities in human liver microsomes. dihydrocapsaicin 70-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 22166891-2 2012 These phytocannabinoids concentration-dependently inhibited S-warfarin 7-hydroxylase and diclofenac 4"-hydroxylase activities of human liver microsomes (HLMs) and recombinant CYP2C9 (rCYP2C9). phytocannabinoids 6-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 22166891-8 2012 These results indicated that the three major phytocannabinoids but not PAHs contained in marijuana smoke potently inhibited CYP2C9 activity and that these cannabinoids can be characterized as direct inhibitors for CYP2C9. phytocannabinoids 45-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 22166891-8 2012 These results indicated that the three major phytocannabinoids but not PAHs contained in marijuana smoke potently inhibited CYP2C9 activity and that these cannabinoids can be characterized as direct inhibitors for CYP2C9. phytocannabinoids 45-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 22166891-8 2012 These results indicated that the three major phytocannabinoids but not PAHs contained in marijuana smoke potently inhibited CYP2C9 activity and that these cannabinoids can be characterized as direct inhibitors for CYP2C9. Cannabinoids 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 22166891-8 2012 These results indicated that the three major phytocannabinoids but not PAHs contained in marijuana smoke potently inhibited CYP2C9 activity and that these cannabinoids can be characterized as direct inhibitors for CYP2C9. Cannabinoids 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 22098100-8 2012 A mechanism-based inhibition of human CYP2C9 by sesamin was recently discovered, suggesting that it is important to evaluate the interaction between sesamin and drugs that are mainly metabolised by CYP2C9. sesamin 48-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22098100-8 2012 A mechanism-based inhibition of human CYP2C9 by sesamin was recently discovered, suggesting that it is important to evaluate the interaction between sesamin and drugs that are mainly metabolised by CYP2C9. sesamin 48-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 22098100-8 2012 A mechanism-based inhibition of human CYP2C9 by sesamin was recently discovered, suggesting that it is important to evaluate the interaction between sesamin and drugs that are mainly metabolised by CYP2C9. sesamin 149-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22098100-8 2012 A mechanism-based inhibition of human CYP2C9 by sesamin was recently discovered, suggesting that it is important to evaluate the interaction between sesamin and drugs that are mainly metabolised by CYP2C9. sesamin 149-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 22927772-1 2012 BACKGROUND: The polymorphisms of VKORC1 and CYP2C9 play increasingly important roles in the inter-individual variability in warfarin dose. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 22927772-10 2012 CONCLUSION: Based on CYP2C9 and VKORC1 genotypes, the pharmacogenetic-based warfarin-dosing algorithm may shorten the time elapse from initiation of warfarin therapy until warfarin maintenance dose. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 22927772-10 2012 CONCLUSION: Based on CYP2C9 and VKORC1 genotypes, the pharmacogenetic-based warfarin-dosing algorithm may shorten the time elapse from initiation of warfarin therapy until warfarin maintenance dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 22927772-10 2012 CONCLUSION: Based on CYP2C9 and VKORC1 genotypes, the pharmacogenetic-based warfarin-dosing algorithm may shorten the time elapse from initiation of warfarin therapy until warfarin maintenance dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 22952875-0 2012 Influence of CYP2C9 and VKORC1 on patient response to warfarin: a systematic review and meta-analysis. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 23353843-2 2012 This cytochrome P450 is considered to be a counterpart of human CYP2C9, which metabolizes many drugs, including diclofenac, ibuprofen or warfarin. Diclofenac 112-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 23353843-2 2012 This cytochrome P450 is considered to be a counterpart of human CYP2C9, which metabolizes many drugs, including diclofenac, ibuprofen or warfarin. Ibuprofen 124-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 23353843-2 2012 This cytochrome P450 is considered to be a counterpart of human CYP2C9, which metabolizes many drugs, including diclofenac, ibuprofen or warfarin. Warfarin 137-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 23353843-9 2012 CONCLUSION: The results presented open the possibility that in humans, rosuvastatin may affect the metabolism of many drugs by influencing expression and activity of CYP2C6 (counterpart of human CYP2C9). Rosuvastatin Calcium 71-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 22393834-1 2012 BACKGROUND: VKORC1, CYP2C9 and CYP4F2 are three critical genes associated with inter-individual variation of warfarin dose. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 22952875-3 2012 Two genes, CYP2C9 and VKORC1, have been associated with this variability, leading to genotype-guided dosing tables in warfarin labeling. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 21726410-0 2011 Effects of the CYP2C9*1/*13 genotype on the pharmacokinetics of lornoxicam. lornoxicam 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 22116191-1 2012 A significant proportion of warfarin dose variability is explained by variation in the genotypes of the cytochrome P450 CYP2C9 and the vitamin K epoxide reductase complex, VKORC1, enzymes that influence warfarin metabolism and sensitivity, respectively. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 22116191-1 2012 A significant proportion of warfarin dose variability is explained by variation in the genotypes of the cytochrome P450 CYP2C9 and the vitamin K epoxide reductase complex, VKORC1, enzymes that influence warfarin metabolism and sensitivity, respectively. Warfarin 203-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 22178823-6 2011 CONCLUSION: It is of important clinical significance in individualized warfarin therapy to systematically study distribution of mutation frequencies at 12 polymorphisms loci in 4 genes including CYP2C9, CYP3A4 , VKORC1 and GGCX related to warfarin pharmacodynamics and pharmacokinetics in the Chinese Han population receiving valve replacement. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 22178823-6 2011 CONCLUSION: It is of important clinical significance in individualized warfarin therapy to systematically study distribution of mutation frequencies at 12 polymorphisms loci in 4 genes including CYP2C9, CYP3A4 , VKORC1 and GGCX related to warfarin pharmacodynamics and pharmacokinetics in the Chinese Han population receiving valve replacement. Warfarin 242-250 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 21726410-1 2011 Lornoxicam is extensively metabolized by CYP2C9, and a CYP2C9*13 is one of the principal variant alleles in East Asian populations. lornoxicam 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 21726410-1 2011 Lornoxicam is extensively metabolized by CYP2C9, and a CYP2C9*13 is one of the principal variant alleles in East Asian populations. lornoxicam 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21726410-2 2011 The aim of this study was to evaluate the effects of CYP2C9*1/*13 on the pharmacokinetic parameters of lornoxicam in healthy individuals. lornoxicam 103-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 21726410-5 2011 In individuals with CYP2C9*1/*13, lornoxicam had a higher C(max) (p < 0.001), a longer half-life (p < 0.001), a lower oral clearance (p < 0.001) and a higher area under the plasma concentration-time curve from zero to infinity (AUC(inf) ) than in CYP2C9*1/*1 individuals (p < 0.001). lornoxicam 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 21726410-5 2011 In individuals with CYP2C9*1/*13, lornoxicam had a higher C(max) (p < 0.001), a longer half-life (p < 0.001), a lower oral clearance (p < 0.001) and a higher area under the plasma concentration-time curve from zero to infinity (AUC(inf) ) than in CYP2C9*1/*1 individuals (p < 0.001). lornoxicam 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 256-262 21726410-6 2011 The C(max) and AUC(inf) of 5"-hydroxylornoxicam were lower in CYP2C9*1/*13 individuals than in CYP2C9*1/*1 individuals, but the half-life of 5"-hydroxylornoxicam did not differ between the two groups. 5'-hydroxylornoxicam 27-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 21726410-6 2011 The C(max) and AUC(inf) of 5"-hydroxylornoxicam were lower in CYP2C9*1/*13 individuals than in CYP2C9*1/*1 individuals, but the half-life of 5"-hydroxylornoxicam did not differ between the two groups. 5'-hydroxylornoxicam 27-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 21726410-7 2011 The half-life, oral clearance and AUC(inf) of lornoxicam were similar in individuals with CYP2C9*1/*13 and those with CYP2C9*1/*3. lornoxicam 46-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 21726410-9 2011 A CYP2C9*1/*13 genotype markedly reduced the conversion of lornoxicam to 5"-hydroxylornoxicam, to a similar extent as that observed with the CYP2C9*1/*3 genotype. lornoxicam 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 2-8 21726410-9 2011 A CYP2C9*1/*13 genotype markedly reduced the conversion of lornoxicam to 5"-hydroxylornoxicam, to a similar extent as that observed with the CYP2C9*1/*3 genotype. lornoxicam 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 21726410-9 2011 A CYP2C9*1/*13 genotype markedly reduced the conversion of lornoxicam to 5"-hydroxylornoxicam, to a similar extent as that observed with the CYP2C9*1/*3 genotype. 5'-hydroxylornoxicam 73-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 2-8 21726410-9 2011 A CYP2C9*1/*13 genotype markedly reduced the conversion of lornoxicam to 5"-hydroxylornoxicam, to a similar extent as that observed with the CYP2C9*1/*3 genotype. 5'-hydroxylornoxicam 73-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 21691805-8 2011 However, only in the gliclazide group a significant association between CYP2C9 genotype and hypoglycaemic attacks was observed (P = 0.035). Gliclazide 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Nelfinavir 164-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Ritonavir 169-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 21930825-3 2011 Statistically significant induction of CYP1A2 (2.1-, 2.9-, and 2.2-fold), CYP2B6 (1.8-, 2.4-, and 4-fold), and CYP2C9 (1.3-, 1.8-, and 2.6-fold) was observed after NFV, RTV, or RIF treatment, respectively (as expected, CYP2D6 was not induced). Rifampin 177-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 21691805-10 2011 CONCLUSIONS: CYP2C9 polymorphisms leading to decreased enzyme activity show a modest impact on the risk of mild hypoglycaemia attacks during oral antidiabetic treatment, with a significant association in patients treated with gliclazide. Gliclazide 226-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 21713461-5 2011 Wogonin was a potent, competitive inhibitor of CYP1A2 (K (i) = 0.24 muM), and a weak inhibitor of CYP2C19 (IC(50) = 101.10 muM), but was not able to inhibit CYP2C9, CYP2D6, CYP2E1 and CYP3A4 (IC(50) > 200 muM). wogonin 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 22116286-0 2011 Inhibition of CYP3A4 and CYP2C9 by podophyllotoxin: implication for clinical drug-drug interactions. Podophyllotoxin 35-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 22116286-3 2011 Inhibition of CYP3A4, CYP2C9, CYP2C8, CYP2D6, CYP2E1 and CYP2A6 by PPT was investigated in the human liver microsomal system. Podophyllotoxin 67-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 22116286-5 2011 The results showed that PPT strongly exhibited inhibitory effects on CYP3A4 and CYP2C9 in a concentration-dependent manner. Podophyllotoxin 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 22116286-7 2011 Inhibition kinetic analysis showed that PPT exhibited competitive inhibition towards CYP3A4 and CYP2C9 with Ki of 1.6 and 2.0 meu M, respectively. Podophyllotoxin 40-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 22116286-9 2011 Our experimental data indicate that potential drug-drug interaction (DDI) might exist when PPT is co-administered with the substrates which mainly undergo CYP3A4- or CYP2C9-mediated metabolism. Podophyllotoxin 91-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 21939641-3 2011 In this brief review, we discussed the impact of CYP2C polymorphisms on the metabolic fate of small-molecule antidiabetics including sulfonylureas, meglitinides, thiazolidinediones, gliptins, and gliflozins, with the key drug-protein molecular interactions highlighted. Sulfonylurea Compounds 133-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-54 22148003-9 2011 In conclusion, fluvastatin should be used with caution in patients with liver cirrhosis, especially with other medications metabolized with CYP2C9. Fluvastatin 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 22338910-0 2011 An instructive case suggesting warfarin resistance which is independent on the regulation of the CYP2C9 and VKORC1 genotype. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 22161100-5 2011 RESULTS: Of the 5 genes, VKORC1, CYP2C9 and GGCX were associated with warfarin stable dose. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 22161100-6 2011 The multiple linear regression analysis indicated that VKORC1, CYP2C9 and GGCX genes, age and weight, had significant influence on inter-individual variation in warfarin stable dose, which contributed 30.2%, 22.8%, 1.5%, 4.7% and 6.7% respectively. Warfarin 163-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 21782804-0 2011 Low dose requirement for warfarin treatment in a patient with CYP2C9*3/*13 genotype. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 21782804-2 2011 Here we describe a patient who was intolerant of warfarin treatment because of impaired drug metabolism related to the CYP2C9*3/*13 genotype and high warfarin sensitivity associated with the VKORC1 1173TT genotype. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 21782804-6 2011 CONCLUSIONS: To the best of our knowledge, this is the first report of a patient with CYP2C9*3/*13 and VKORC1 1173TT genotypes who had slower than normal warfarin metabolism, resulting in the need to administer an extremely low dose of warfarin in order to achieve the target INR value. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 21782804-6 2011 CONCLUSIONS: To the best of our knowledge, this is the first report of a patient with CYP2C9*3/*13 and VKORC1 1173TT genotypes who had slower than normal warfarin metabolism, resulting in the need to administer an extremely low dose of warfarin in order to achieve the target INR value. Warfarin 236-244 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 21143257-1 2011 WHAT IS KNOWN AND OBJECTIVE: Miconazole is a strong inhibitor of CYP2C9, one of the main enzymes involved in the metabolism of warfarin. Miconazole 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 21143257-1 2011 WHAT IS KNOWN AND OBJECTIVE: Miconazole is a strong inhibitor of CYP2C9, one of the main enzymes involved in the metabolism of warfarin. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 22122181-4 2011 The genes encoding for cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) are the major genetic determinants of warfarin pharmacokinetics and pharmacodynamics, respectively. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-48 22122181-4 2011 The genes encoding for cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) are the major genetic determinants of warfarin pharmacokinetics and pharmacodynamics, respectively. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 22122181-6 2011 The CYP2C9 gene has also been associated with bleeding risk with warfarin. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21939641-3 2011 In this brief review, we discussed the impact of CYP2C polymorphisms on the metabolic fate of small-molecule antidiabetics including sulfonylureas, meglitinides, thiazolidinediones, gliptins, and gliflozins, with the key drug-protein molecular interactions highlighted. meglitinide 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-54 21939641-3 2011 In this brief review, we discussed the impact of CYP2C polymorphisms on the metabolic fate of small-molecule antidiabetics including sulfonylureas, meglitinides, thiazolidinediones, gliptins, and gliflozins, with the key drug-protein molecular interactions highlighted. Thiazolidinediones 162-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-54 22023129-7 2011 For all 3 species, inhibitors of CYP3A4, CYP2A6, CYP2C19, CYP2B6, and CYP2C9 diminished N-demethylation of ketamine. Ketamine 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 21964418-4 2011 Although all of the oxidation reactions tested were more or less inhibited by PFOS, diclofenac 4"-hydroxylation mediated mainly by CYP2C9 was most strongly inhibited (K(i) value of 40 nM), followed by paclitaxel 6alpha-hydroxylation mediated mainly by CYP2C8 (K(i) value of 4 muM). diclofenac 4" 84-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 21964418-8 2011 These results suggest that the metabolism of medicines which are substrates for CYP2C9 may be altered by PFOS in human bodies, and that PFOS is a mechanism-based inhibitor of CYP2A6. perfluorooctane sulfonic acid 105-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21964418-8 2011 These results suggest that the metabolism of medicines which are substrates for CYP2C9 may be altered by PFOS in human bodies, and that PFOS is a mechanism-based inhibitor of CYP2A6. perfluorooctane sulfonic acid 136-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 22023129-7 2011 For all 3 species, inhibitors of CYP3A4, CYP2A6, CYP2C19, CYP2B6, and CYP2C9 diminished N-demethylation of ketamine. Nitrogen 88-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 22023129-8 2011 Anti-CYP3A4, anti-CYP2C9, and anti-CYP2B6 antibodies also inhibited ketamine N-demethylation. Ketamine 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 22023129-11 2011 Although the human CYP2C9 inhibitor blocked ketamine N-demethylation completely in the canine ortholog CYP2C21, a strong inhibition was also obtained by the chemical inhibitors of CYP2C19 and CYP2B6. Ketamine 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 22023129-13 2011 CONCLUSIONS AND CLINICAL RELEVANCE: Human-specific inhibitors of CYP2A6, CYP2C19, CYP3A4, CYP2B6, and CYP2C9 diminished ketamine N-demethylation in dogs and horses. ketamine n 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 21953762-5 2011 Naringenin was able to inhibit CYP19, CYP2C9, and CYP2C19 with IC50 values below 5 muM. naringenin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22152281-3 2011 Unlike other statins, the cyclopropyl group on the pitavastatin molecule appears to divert the drug away from metabolism by cytochrome P450 (CYP) 3 A4 and allows only a small degree of clinically insignificant metabolism by CYP2C9. pitavastatin 51-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-230 21968795-1 2011 This short communication is aimed to investigate whether the widely used hypolipidemic drug fenofibrate affects CYP2C11 and CYP2C6 in rats, both counterparts of human CYP2C9, known to metabolise many drugs including S-warfarin and largely used non-steroidal antiinflammatory drugs such as ibuprofen, diclofenac and others. Fenofibrate 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 21968795-1 2011 This short communication is aimed to investigate whether the widely used hypolipidemic drug fenofibrate affects CYP2C11 and CYP2C6 in rats, both counterparts of human CYP2C9, known to metabolise many drugs including S-warfarin and largely used non-steroidal antiinflammatory drugs such as ibuprofen, diclofenac and others. Sulfur 216-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 21968795-5 2011 The results indicate that the combination of fenofibrate with drugs metabolised by CYP2C9 in humans should be taken with caution as it may lead, for example, to the potentiation of warfarin effects. Warfarin 181-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 21953762-7 2011 Whereas (S)-naringenin was 2-fold more potent as an inhibitor of CYP19 and CYP2C19 than (R)-naringenin, (R)-naringenin was 2-fold more potent for CYP2C9 and CYP3A. (R)-naringenin 104-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 21590310-2 2011 We designed a study to determine the associations between the polymorphisms and haplotype structures of CYP2C9 and VKORC1 and warfarin dose response in Sudanese patients, one of the most genetically diverse populations in Africa. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 21590310-4 2011 RESULTS: Patients with the CYP2C9*2,*5,*6, or *11 variant required a daily warfarin dose that was 21% lower than those with CYP2C9*1/*1 (4.7 vs 5.8 mg/day, P < 0.001). Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 21590310-7 2011 CONCLUSION: Our data revealed that VKORC1 and CYP2C9 polymorphisms are important factors that influence warfarin dose response in Sudanese patients. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21651319-1 2011 BACKGROUND: Polymorphisms in the genes encoding the cytochrome P450 2C9 enzyme (CYP2C9) and the vitamin K epoxide reductase (VKORC1) are known to contribute to variability in sensitivity to coumarins. Coumarins 190-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 22012312-1 2011 A significant proportion of the interindividual variability in warfarin dose requirements can be explained on the basis of CYP2C9 and VKORC1 genotypes. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 21605066-0 2011 Comparison of metabolic soft spot predictions of CYP3A4, CYP2C9 and CYP2D6 substrates using MetaSite and StarDrop. stardrop 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 21651319-1 2011 BACKGROUND: Polymorphisms in the genes encoding the cytochrome P450 2C9 enzyme (CYP2C9) and the vitamin K epoxide reductase (VKORC1) are known to contribute to variability in sensitivity to coumarins. Coumarins 190-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21651319-2 2011 Patients with certain common genetic variants of CYP2C9 (*2 & *3) or a VKORC1 polymorphism (-1639A Allele) require a lower dose of coumarin and are also at higher risk for over-anticoagulation and serious bleeding. coumarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 21550074-10 2011 With specific inhibitors of CYP3A, CYP2B6, CYP2D6, CYP2C9, and CYP2C19, active metabolite production was decreased by 38% +- 15% with 4-(4-chlorobenzyl)pyridine (CYP2B6 inhibitor) and by 45 +- 16% with ketoconazole (CYP3A inhibitor). 4-(4-Chlorobenzyl)pyridine 134-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 22011696-5 2011 DISCUSSION: The drug interaction between warfarin and S-1 presumably leads to elevated PT-INR because the 5-fluorouracil (5-FU), which is metabolite of FT in S-1, inhibits the metabolic processing of S-warfarin by cytochrome P450 (CYP) 2C9. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-239 22011696-5 2011 DISCUSSION: The drug interaction between warfarin and S-1 presumably leads to elevated PT-INR because the 5-fluorouracil (5-FU), which is metabolite of FT in S-1, inhibits the metabolic processing of S-warfarin by cytochrome P450 (CYP) 2C9. Fluorouracil 106-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-239 22011696-5 2011 DISCUSSION: The drug interaction between warfarin and S-1 presumably leads to elevated PT-INR because the 5-fluorouracil (5-FU), which is metabolite of FT in S-1, inhibits the metabolic processing of S-warfarin by cytochrome P450 (CYP) 2C9. Fluorouracil 122-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-239 21550074-10 2011 With specific inhibitors of CYP3A, CYP2B6, CYP2D6, CYP2C9, and CYP2C19, active metabolite production was decreased by 38% +- 15% with 4-(4-chlorobenzyl)pyridine (CYP2B6 inhibitor) and by 45 +- 16% with ketoconazole (CYP3A inhibitor). Ketoconazole 202-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 22120087-5 2011 The results dis-played that three variants of the warfarin metabolism-related genetic polymorphism (CYP2C9*2, CYP2C9*3, and VKORC1(-1693)) could be simultaneously detected using three different sequencing primers in a single-tube (one test), and 96 tests could be carried out each time. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 21713378-3 2011 In this study, we evaluated allele frequencies and effects of CYP2C9 and VKORC1 on warfarin response during initial anticoagulation therapy in Korean patients. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 21713378-10 2011 Patients with the CYP2C9*3 allele received a lower warfarin dose (P = 0.018) and tended to show more rapid PT-INR increase than CYP2C9*1/*1 genotype. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 21713378-12 2011 The CYP2C9*3 and VKORC1-1639G alleles influenced warfarin response during the first month of anticoagulation therapy. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21713378-13 2011 Considering these results, CYP2C9 and VKORC1 genotyping can be an useful tool to estimate initial warfarin dose and frequency of PT-INR monitoring during the first month of anticoagulation therapy. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 22120087-5 2011 The results dis-played that three variants of the warfarin metabolism-related genetic polymorphism (CYP2C9*2, CYP2C9*3, and VKORC1(-1693)) could be simultaneously detected using three different sequencing primers in a single-tube (one test), and 96 tests could be carried out each time. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 21841812-0 2011 Frequency of CYP2C9 alleles in Koreans and their effects on losartan pharmacokinetics. Losartan 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 21745145-5 2011 Of the CYPs evaluated, NGF1568 was found to inhibit only CYP2C9, by an uncompetitive mechanism and with a K(i) value of 349 muM. NGF 1568 23-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 21771587-2 2011 Herein, we report a case study in which the plasma concentration of the CYP2C9 substrate (S)-warfarin, and not the CYP3A4 substrate (R)-warfarin, increased with itraconazole coadministration. (S)-Warfarin 89-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21771587-2 2011 Herein, we report a case study in which the plasma concentration of the CYP2C9 substrate (S)-warfarin, and not the CYP3A4 substrate (R)-warfarin, increased with itraconazole coadministration. Itraconazole 161-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21744854-0 2011 Membrane position of ibuprofen agrees with suggested access path entrance to cytochrome P450 2C9 active site. Ibuprofen 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-96 21744854-4 2011 We constructed an atomistic model of complete CYP2C9 in a dioleoylphosphatidylcholine membrane and evolved it by molecular dynamics simulations in explicit water on a 100+ ns time-scale. 1,2-oleoylphosphatidylcholine 58-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21744854-7 2011 The positions of openings of the substrate access and product egress channels correspond to free energy minima of CYP2C9 substrate ibuprofen and its metabolite in the membrane, respectively. Ibuprofen 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 21757329-0 2011 Polysaccharide peptides from Coriolus versicolor competitively inhibit tolbutamide 4-hydroxylation in specific human CYP2C9 isoform and pooled human liver microsomes. Polysaccharides 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 21757329-0 2011 Polysaccharide peptides from Coriolus versicolor competitively inhibit tolbutamide 4-hydroxylation in specific human CYP2C9 isoform and pooled human liver microsomes. Tolbutamide 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 21841812-5 2011 The enzymatic activity of each CYP2C9 genotype was evaluated using losartan as the substrate. Losartan 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 21841812-8 2011 In the pharmacokinetics studies, the AUC(0- ) of losartan in CYP2C9*3/*3 subjects was 1.42-fold larger than that in CYP2C9*1/*1 subjects, and the AUC(0- ) of E-3174, a more active metabolite of losartan, in CYP2C9*3/*3 subjects was only 12% of that in CYP2C9*1/*1 subjects. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21841812-8 2011 In the pharmacokinetics studies, the AUC(0- ) of losartan in CYP2C9*3/*3 subjects was 1.42-fold larger than that in CYP2C9*1/*1 subjects, and the AUC(0- ) of E-3174, a more active metabolite of losartan, in CYP2C9*3/*3 subjects was only 12% of that in CYP2C9*1/*1 subjects. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 21841812-8 2011 In the pharmacokinetics studies, the AUC(0- ) of losartan in CYP2C9*3/*3 subjects was 1.42-fold larger than that in CYP2C9*1/*1 subjects, and the AUC(0- ) of E-3174, a more active metabolite of losartan, in CYP2C9*3/*3 subjects was only 12% of that in CYP2C9*1/*1 subjects. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 21900891-2 2011 Common genetic variants in the cytochrome P450-2C9 (CYP2C9) and vitamin K-epoxide reductase complex (VKORC1) enzymes, in addition to known nongenetic factors, account for ~50% of warfarin dose variability. Warfarin 179-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-50 21841812-8 2011 In the pharmacokinetics studies, the AUC(0- ) of losartan in CYP2C9*3/*3 subjects was 1.42-fold larger than that in CYP2C9*1/*1 subjects, and the AUC(0- ) of E-3174, a more active metabolite of losartan, in CYP2C9*3/*3 subjects was only 12% of that in CYP2C9*1/*1 subjects. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 21841812-10 2011 CYP2C9*3/*3 subjects metabolized much less losartan into E-3174 than CYP2C9*1/*1 subjects. Losartan 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21841812-10 2011 CYP2C9*3/*3 subjects metabolized much less losartan into E-3174 than CYP2C9*1/*1 subjects. losartan carboxylic acid 57-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21966608-0 2011 Polymorphisms of Aspirin-Metabolizing Enzymes CYP2C9, NAT2 and UGT1A6 in Aspirin-Intolerant Urticaria. Aspirin 17-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21966608-0 2011 Polymorphisms of Aspirin-Metabolizing Enzymes CYP2C9, NAT2 and UGT1A6 in Aspirin-Intolerant Urticaria. Aspirin 73-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21966608-1 2011 Acetyl salicylic acid (ASA) is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6), cytochrome P4502C9 (CYP2C9), and N-acetyl transferase 2 (NAT2). Aspirin 0-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-106 21966608-1 2011 Acetyl salicylic acid (ASA) is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6), cytochrome P4502C9 (CYP2C9), and N-acetyl transferase 2 (NAT2). Aspirin 0-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 21966608-1 2011 Acetyl salicylic acid (ASA) is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6), cytochrome P4502C9 (CYP2C9), and N-acetyl transferase 2 (NAT2). Aspirin 23-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-106 21966608-1 2011 Acetyl salicylic acid (ASA) is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6), cytochrome P4502C9 (CYP2C9), and N-acetyl transferase 2 (NAT2). Aspirin 23-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 21981797-1 2011 BACKGROUND: The pharmacokinetics and pharmacodynamics of warfarin are affected by polymorphisms in the genes coding for cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1). Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-139 21981797-1 2011 BACKGROUND: The pharmacokinetics and pharmacodynamics of warfarin are affected by polymorphisms in the genes coding for cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1). Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 21981797-5 2011 RESULTS: A multivariate regression model including the variables of age, VKORC1 and CYP2C9 genotype, body surface area, and statin status produced the best model for estimating the warfarin dose (R(2) = 0.62). Warfarin 181-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 21900891-2 2011 Common genetic variants in the cytochrome P450-2C9 (CYP2C9) and vitamin K-epoxide reductase complex (VKORC1) enzymes, in addition to known nongenetic factors, account for ~50% of warfarin dose variability. Warfarin 179-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 21900891-3 2011 The purpose of this article is to assist in the interpretation and use of CYP2C9 and VKORC1 genotype data for estimating therapeutic warfarin dose to achieve an INR of 2-3, should genotype results be available to the clinician. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 21148049-9 2011 The concentrations of R- and S-warfarin in plasma were significantly correlated with CYP2C9 genotypes. r- and s-warfarin 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24250426-2 2011 Diclofenac liver cytotoxicity was associated with reactive oxygen species (ROS) formation and lipid peroxidation which were inhibited by antioxidants and ROS scavengers, ferric chelator, inhibitors of reduced CYP2E1 and CYP2C9, mitochondrial permeability transition (MPT) pore sealing agents and endocytosis inhibitors. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 24250426-8 2011 We finally concluded that diclofenac hepatotoxicity is a result of metabolic activation by CYP2E1 and CYP2C9 and ROS formation, leading to a mitochondrial/lysosomal toxic cross-talk in the liver hepatocytes. Diclofenac 26-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 21148049-10 2011 For acenocoumarol, time to reach target INR was more prolonged in patients carrying any CYP2C9 variant allele but failed to reach statistical significance because of low numbers of patients. Acenocoumarol 4-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 21148049-13 2011 The authors showed that both CYP2C9 and VKORC1 polymorphisms are common in Lebanon and influence warfarin and acenocoumarol dose requirements, with the CYP2C9*2 polymorphism having less effect on acenocoumarol, the most commonly used oral anticoagulant in Lebanon. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 21148049-13 2011 The authors showed that both CYP2C9 and VKORC1 polymorphisms are common in Lebanon and influence warfarin and acenocoumarol dose requirements, with the CYP2C9*2 polymorphism having less effect on acenocoumarol, the most commonly used oral anticoagulant in Lebanon. Acenocoumarol 110-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 22321278-0 2011 [Impact of CYP2C9 and VKORC1 polymorphism on warfarin response during initiation of therapy]. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 21638223-0 2011 Extreme sensitivity to acenocoumarol therapy in patient with both VKORC.-1639 A/A and CYP2C9*1/*3 genotypes. Acenocoumarol 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 21638223-8 2011 The genotyping results performed after introducing of VKA showed the patient was homozygous for vitamin K epoxide reductase (VKORC1) c.-1639 G>A, and heterozygous for cytochrome P450 2C9 (CYP2C9)*3 which pointed to extreme sensitivity to acenocoumarol. Acenocoumarol 241-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-189 21726172-8 2011 Although clinical doses of toremifene produce steady state exposures to toremifene and NDMT that may be sufficient to cause pharmacokinetic drug-drug interactions with other drugs metabolised by CYP2B6, CYP2C8, CYP3A4, CYP2C9 and CYP2C19, these data indicate that toremifene is unlikely to play a role in clinical drug-drug interactions with substrate drugs of CYP1A2 and CYP2D6. Toremifene 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 22321278-1 2011 OBJECTIVE: To investigate potential contributions of genetic variants of cytochrome P-450 2C9 (CYP2C9) and vitamin K expoxide reductase (VKORC1) to the anticoagulation response during the initiation of warfarin therapy in the Han Chinese population. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-93 22321278-1 2011 OBJECTIVE: To investigate potential contributions of genetic variants of cytochrome P-450 2C9 (CYP2C9) and vitamin K expoxide reductase (VKORC1) to the anticoagulation response during the initiation of warfarin therapy in the Han Chinese population. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 22321278-10 2011 CONCLUSION: Detecting genetic polymorphism of CYP2C9 and VKORC1 could guide clinical use of warfarin to reduce the risk of adverse reactions including bleeding in patients receiving chronic anticoagulation therapy. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21320153-1 2011 AIMS: To investigate the influence of polymorphisms in CYP2C9, VKORC1, CYP4F2 and F2 genes on warfarin dose-response and develop a model including genetic and non-genetic factors for warfarin dose prediction needed for each patient. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21725053-1 2011 Single-nucleotide polymorphisms in genes that affect warfarin metabolism (cytochrome P450 2C9 gene, CYP2C9) and response (vitamin K epoxide reductase complex 1 gene, VKORC1) have an important influence on warfarin therapy, particularly during initiation; however, there is a lack of consensus regarding the optimal pharmacogenetics-based initiation strategy. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-93 21725053-1 2011 Single-nucleotide polymorphisms in genes that affect warfarin metabolism (cytochrome P450 2C9 gene, CYP2C9) and response (vitamin K epoxide reductase complex 1 gene, VKORC1) have an important influence on warfarin therapy, particularly during initiation; however, there is a lack of consensus regarding the optimal pharmacogenetics-based initiation strategy. Warfarin 205-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-93 21725053-1 2011 Single-nucleotide polymorphisms in genes that affect warfarin metabolism (cytochrome P450 2C9 gene, CYP2C9) and response (vitamin K epoxide reductase complex 1 gene, VKORC1) have an important influence on warfarin therapy, particularly during initiation; however, there is a lack of consensus regarding the optimal pharmacogenetics-based initiation strategy. Warfarin 205-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 21622626-1 2011 Our previous study revealed that CYP2C9 played a central role in sesamin monocatecholization. sesamin 65-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 21622626-14 2011 On the basis of these results, we concluded that CYP2C9, UGT2B7, and COMT played essential roles in the metabolism of sesamin in the human liver. sesamin 118-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 21338443-0 2011 A candidate gene study of antiepileptic drug tolerability and efficacy identifies an association of CYP2C9 variants with phenytoin toxicity. Phenytoin 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 21338443-8 2011 RESULTS: We identified significant associations of CYP2C9 variant alleles with presence of phenytoin (PHT) adverse drug reactions (ADRs) and of GSTM1 copy number variation with the presence of carbamazepine ADRs. Phenytoin 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 21338443-8 2011 RESULTS: We identified significant associations of CYP2C9 variant alleles with presence of phenytoin (PHT) adverse drug reactions (ADRs) and of GSTM1 copy number variation with the presence of carbamazepine ADRs. Phenytoin 104-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 21826053-2 2011 The inhibitory potential of corydaline on the activities of seven major human cytochrome P450 and four UDP-glucuronosyltransferase enzymes in human liver microsomes was investigated using LC-tandem MS. Corydaline was found to inhibit CYP2C19-catalyzed S-mephenytoin-4"-hydroxylatoin and CYP2C9-catalyzed diclofenac 4-hydroxylation, with K(i) values of 1.7 and 7.0 mM, respectively. corydaline 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 287-293 22040439-0 2011 Extremely low warfarin dose in patients with genotypes of CYP2C9*3/*3 and VKORC1-1639A/A. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 22040439-1 2011 BACKGROUND: Patients with the genotypes of both CYP2C9*3/*3 and VKORC1-1639 A/A are expected to require the lowest dose of warfarin, and to have a greatly increased risk of bleeding. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 22040439-13 2011 CONCLUSIONS: Two Chinese with the rare genotypes of both CYP2C9*3/*3 and VKORC1-1639 A/A were found to require the extremely low dose of warfarin. Warfarin 137-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 22040439-14 2011 The pharmacogenetic algorithm incorporating the variances of VKORC1 and CYP2C9 genotypes, as well as the non-genetic factors could predict their stable dose of warfarin with high accuracy. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 21906462-4 2011 TAM is known to be metabolised to its active metabolites N-desmethyl TAM and 4-hydroxytamoxifen by a number of CYP450 enzymes, including CYP2D6, CYP3A4, CYP2C9, CYP2C19 and CYP2B6. Tamoxifen 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 21906462-4 2011 TAM is known to be metabolised to its active metabolites N-desmethyl TAM and 4-hydroxytamoxifen by a number of CYP450 enzymes, including CYP2D6, CYP3A4, CYP2C9, CYP2C19 and CYP2B6. hydroxytamoxifen 77-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 21985811-11 2011 INTERPRETATION & CONCLUSIONS: Our results demonstrated significant involvement of CYP2C9 genetic variants in the modulation of epilepsy pharmacotherapy confirming the important role of CYP2C9 mutants preventing epilepsy patients from developing drug resistance. Adenosine Monophosphate 16-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 21386709-6 2011 Solid evidence supports the importance of gene variants in CYP2C9 and VKORC1 for warfarin dosing and in CYP2C19 for clopidogrel response in adult patients. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 21476064-0 2011 Population pharmacokinetic analysis of glimepiride with CYP2C9 genetic polymorphism in healthy Korean subjects. glimepiride 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 21476064-1 2011 PURPOSE: The purpose of this study was to develop a population pharmacokinetic (PPK) model of glimepiride and to investigate the influence of genetic polymorphisms in CYP2C9 on the PPK of glimepiride in healthy Korean subjects. glimepiride 188-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 21476064-6 2011 The CYP2C9 genotypes as covariate significantly (P < 0.001) influenced the apparent oral clearance (CL/F) of glimepiride. glimepiride 112-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21476064-7 2011 The estimated CL/F of glimepiride was higher (1.60-fold) in CYP2C9*1/*1 subjects than in CYP2C9*1/*3 subjects. glimepiride 22-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 21476064-7 2011 The estimated CL/F of glimepiride was higher (1.60-fold) in CYP2C9*1/*1 subjects than in CYP2C9*1/*3 subjects. glimepiride 22-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 21476064-8 2011 CONCLUSIONS: This study indicates that genetic polymorphisms of CYP2C9 influence the substantial interindividual variability in the disposition of glimepiride, and these polymorphisms may affect the clinical response to glimepiride therapy. glimepiride 147-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 21476064-8 2011 CONCLUSIONS: This study indicates that genetic polymorphisms of CYP2C9 influence the substantial interindividual variability in the disposition of glimepiride, and these polymorphisms may affect the clinical response to glimepiride therapy. glimepiride 220-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 21516380-0 2011 Clinical pharmacokinetics of ketoprofen enantiomers in wild type of Cyp 2c8 and Cyp 2c9 patients with rheumatoid arthritis. Ketoprofen 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-87 21826053-2 2011 The inhibitory potential of corydaline on the activities of seven major human cytochrome P450 and four UDP-glucuronosyltransferase enzymes in human liver microsomes was investigated using LC-tandem MS. Corydaline was found to inhibit CYP2C19-catalyzed S-mephenytoin-4"-hydroxylatoin and CYP2C9-catalyzed diclofenac 4-hydroxylation, with K(i) values of 1.7 and 7.0 mM, respectively. corydaline 202-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 287-293 21826053-5 2011 These in vitro results suggest that corydaline should be evaluated for potential pharmacokinetic drug interactions in vivo due to potent inhibition of CYP2C19 and CYP2C9. corydaline 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 21332946-0 2011 Effect of atorvastatin on CYP2C9 metabolic activity as measured by the formation rate of losartan metabolite in hypercholesterolaemic patients. Atorvastatin 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 21332946-0 2011 Effect of atorvastatin on CYP2C9 metabolic activity as measured by the formation rate of losartan metabolite in hypercholesterolaemic patients. Losartan 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 21332946-4 2011 The aim of this study was to determine the effect of atorvastatin on the activity of CYP2C9 in a group of Turkish hypercholesterolaemic patients. Atorvastatin 53-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21332946-5 2011 We prospectively investigated the atorvastatin effect on CYP2C9 activity in a sample of Turkish hypercholesterolaemia patients (11 women, 7 men) who commenced atorvastatin (10 mg/day). Atorvastatin 34-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 21332946-6 2011 Losartan was used as a probe drug to determine CYP2C9 metabolic activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 21636598-1 2011 AIMS: Polymorphisms in CYP2C9 and VKORC1 influence patients" phenprocoumon (PHE) and acenocoumarol (ACE) dose requirements. Phenprocoumon 61-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 21636598-1 2011 AIMS: Polymorphisms in CYP2C9 and VKORC1 influence patients" phenprocoumon (PHE) and acenocoumarol (ACE) dose requirements. Phenprocoumon 76-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 21636598-1 2011 AIMS: Polymorphisms in CYP2C9 and VKORC1 influence patients" phenprocoumon (PHE) and acenocoumarol (ACE) dose requirements. Acenocoumarol 85-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 21636598-1 2011 AIMS: Polymorphisms in CYP2C9 and VKORC1 influence patients" phenprocoumon (PHE) and acenocoumarol (ACE) dose requirements. Acenocoumarol 100-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 21636598-4 2011 Single nucleotide polymorphisms relevant to coumarin derivative dosing on the CYP2C9 and VKORC1 genes were determined. coumarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 21336994-0 2011 Influence of CYP2C9 gene polymorphisms on response to glibenclamide in type 2 diabetes mellitus patients. Glyburide 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 21636598-8 2011 CYP2C9 and VKORC1 genotype, weight, height, sex, age, and amiodarone use contributed to the maintenance dose of PHE and ACE. Phenprocoumon 112-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21336994-1 2011 PURPOSE: The antidiabetic drug glibenclamide is metabolized by the enzyme cytochrome P450 2C9 (CYP2C9) encoded by the polymorphic gene CYP2C9. Glyburide 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-93 21636598-8 2011 CYP2C9 and VKORC1 genotype, weight, height, sex, age, and amiodarone use contributed to the maintenance dose of PHE and ACE. Acenocoumarol 120-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21336994-1 2011 PURPOSE: The antidiabetic drug glibenclamide is metabolized by the enzyme cytochrome P450 2C9 (CYP2C9) encoded by the polymorphic gene CYP2C9. Glyburide 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 22075505-4 2011 The pharmacokinetic sensitivity of warfarin was significantly higher in the CYP2C9 *1/*3 genotypes than in the CYP2C9 *1/*1 genotypes [ratio of S-warfarin concentration/dose, 0.53 vs. 0.21; p=0.01]. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 21336994-1 2011 PURPOSE: The antidiabetic drug glibenclamide is metabolized by the enzyme cytochrome P450 2C9 (CYP2C9) encoded by the polymorphic gene CYP2C9. Glyburide 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 21336994-2 2011 Previous studies involving healthy volunteers have shown a significant influence of variant CYP2C9 genotypes on glibenclamide metabolism. Glyburide 112-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 21336994-3 2011 The aim of this study was to investigate the influence of genetic polymorphisms of CYP2C9 on the response to glibenclamide and on glibenclamide plasma levels in type 2 diabetes mellitus patients. Glyburide 109-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 21336994-9 2011 There was a significant association (p < 0.001) between genotype status of CYP2C9 and the control of diabetes in patients receiving treatment with glibenclamide. Glyburide 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 21336994-11 2011 CONCLUSION: The type 2 diabetes mellitus patients participating in this study with variant genotypes of CYP2C9 were found to respond better to treatment with glibenclamide than those with the normal genotype. Glyburide 158-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 21120554-5 2011 Interestingly, at the binding sites of all PPI-CYP2C9 complexes except for Lan/CYP2C9, there are hydrogen-bonding networks made of PPIs, water molecules, and some residues of 2C9. Hydrogen 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 21120554-5 2011 Interestingly, at the binding sites of all PPI-CYP2C9 complexes except for Lan/CYP2C9, there are hydrogen-bonding networks made of PPIs, water molecules, and some residues of 2C9. Hydrogen 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 21120554-5 2011 Interestingly, at the binding sites of all PPI-CYP2C9 complexes except for Lan/CYP2C9, there are hydrogen-bonding networks made of PPIs, water molecules, and some residues of 2C9. Water 137-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 21120554-5 2011 Interestingly, at the binding sites of all PPI-CYP2C9 complexes except for Lan/CYP2C9, there are hydrogen-bonding networks made of PPIs, water molecules, and some residues of 2C9. Water 137-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 21704641-7 2011 Seven of 14 recombinant human CYP enzymes examined (CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5) were capable of metabolizing CBD. Cannabidiol 142-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 22075505-4 2011 The pharmacokinetic sensitivity of warfarin was significantly higher in the CYP2C9 *1/*3 genotypes than in the CYP2C9 *1/*1 genotypes [ratio of S-warfarin concentration/dose, 0.53 vs. 0.21; p=0.01]. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 22075505-7 2011 The increase of over-anticoagulation risk and warfarin sensitivity is related to the CYP2C9*3 allele and old age with the VKORC1 1173 T allele in Korean patients with thromboembolic disease. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21672908-8 2011 Associations between known variants in VKORC1 and CYP2C9 and warfarin stable weekly dose were performed with linear regression adjusted for age, gender, and body mass index. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 21666702-8 2011 Ticlopidine inhibited CYP2C9 and 3A4 with IC50 values of 26.0 and 32.3 mumol/L, respectively. Ticlopidine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 21666702-10 2011 CONCLUSION: The significant increase in the AUC of losartan (9 mg/kg) by ticlopidine (10 mg/kg) could be attributed to the inhibition of CYP2C9- and 3A4-mediated losartan metabolism in small intestine and/or in liver. Losartan 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21666702-10 2011 CONCLUSION: The significant increase in the AUC of losartan (9 mg/kg) by ticlopidine (10 mg/kg) could be attributed to the inhibition of CYP2C9- and 3A4-mediated losartan metabolism in small intestine and/or in liver. Ticlopidine 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21666702-10 2011 CONCLUSION: The significant increase in the AUC of losartan (9 mg/kg) by ticlopidine (10 mg/kg) could be attributed to the inhibition of CYP2C9- and 3A4-mediated losartan metabolism in small intestine and/or in liver. Losartan 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21460231-7 2011 In addition, PPOH and MS-PPOH displayed time- and NADPH-dependent inhibition of CYP2C9 and other epoxygenases. propenylphosphonic acid 13-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21460231-7 2011 In addition, PPOH and MS-PPOH displayed time- and NADPH-dependent inhibition of CYP2C9 and other epoxygenases. propenylphosphonic acid 25-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21460231-7 2011 In addition, PPOH and MS-PPOH displayed time- and NADPH-dependent inhibition of CYP2C9 and other epoxygenases. NADP 50-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21476972-7 2011 It has been demonstrated that CYP2J2 generates cardioprotective EETs, whereas another isozyme in the heart, CYP2C, generates EETs as well as detrimental ROS. Reactive Oxygen Species 153-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-113 21672908-5 2011 The authors then conducted an association analysis between an automatically extracted stable weekly dose of warfarin and four genetic variants of VKORC1 and CYP2C9 genes. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 21672908-10 2011 Using the automatically extracted weekly doses of warfarin, the authors successfully replicated the previous known associations between warfarin stable dose and genetic variants in VKORC1 and CYP2C9. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21672908-10 2011 Using the automatically extracted weekly doses of warfarin, the authors successfully replicated the previous known associations between warfarin stable dose and genetic variants in VKORC1 and CYP2C9. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21635147-0 2011 Novel CYP2C9 and VKORC1 gene variants associated with warfarin dosage variability in the South African black population. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 21493749-4 2011 We hypothesize that this may occur because of the sex steroid-dependent activation of estrogen receptor alpha (ERalpha) with further transactivation of the CYP2C9 gene. Steroids 54-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 21365364-2 2011 In vitro probe-based high performance liquid chromatography assays were developed to determine CYP2C9-dependent tolbutamide methylhydroxylation, CYP2D6-dependent dextromethorphan O-demethylation and CYP3A4-dependent testosterone 6beta-hydroxylation activities in the presence and absence of AP extracts and andrographolide. Tolbutamide 112-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 21635147-9 2011 CONCLUSION: We demonstrate that CYP2C9*8 and two novel CYP2C9 SNPs (g.16179 and g.46028) are associated with a decrease in warfarin dosage, beta-blockers are independently associated with a decrease in warfarin dosage and two known VKORC1 variants (rs7200749 and rs7294) are associated with an increase in warfarin dosage. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21635147-10 2011 The CYP2C9 and VKORC1 variants and a small subset of environmental factors used in the study explain approximately 45% of warfarin dosage variability in the South African black population. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21639946-1 2011 BACKGROUND: CYP2C9 and VKORC1 are two major genetic factors associated with inter-individual variability in warfarin dose. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 21447389-4 2011 This was done by using human substrates of CYP3A4 (verapamil and testosterone), CYP2C9 (diclofenac) and CYP2D6 (dextromethorphan). Diclofenac 88-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21639946-4 2011 METHODS: Direct sequencing method was used to identify SNPs in CYP2C9, VKORC1, CYP4F2, EPHX1, PROC and GGCX genes in warfarin-treated patients. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21639946-6 2011 RESULTS: From the 40 SNPs analyzed, CYP2C9 rs17847036 and VKORC1 rs9923231 showed significant association with warfarin sensitivity. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 21639946-9 2011 CYP2C9 and VKORC1 genetic variants as well as non-genetic factors such as age, body weight and body height account for 15.4% of variance in warfarin dose among our study population. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21110192-0 2011 Contribution of VKORC1 and CYP2C9 polymorphisms in the interethnic variability of warfarin dose in Malaysian populations. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 21574568-4 2011 We used structure-activity data from the PubChem database to develop a topomer CoMFA model that guided the design of novel sulfonamides with high selectivity for HIF-1 over CYP2C9 inhibition. Sulfonamides 123-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 21356265-0 2011 Reduced catalytic activity of human CYP2C9 natural alleles for gliclazide: molecular dynamics simulation and docking studies. Gliclazide 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 21356265-1 2011 Amongst sulfonylureas, gliclazide is one of the mostly prescribed drugs to diabetic patients and is metabolized extensively by P450 CYP2C9. Sulfonylurea Compounds 8-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 21356265-1 2011 Amongst sulfonylureas, gliclazide is one of the mostly prescribed drugs to diabetic patients and is metabolized extensively by P450 CYP2C9. Gliclazide 23-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 21356265-2 2011 Among 24-CYP2C9 alleles, the *2/*2 and *3/*3 genotypes showed significantly lower gliclazide clearances with reductions of 25 and 57%, respectively. Gliclazide 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 21356265-4 2011 In the present study, we used molecular dynamics simulation and autodocking studies to provide models for gliclazide-bound complexes of CYP2C9*2, *3 and *2/*3 mutants, which give insight into CYP2C9-gliclazide interactions and explain the reduced enzymatic activity seen in these variants. Gliclazide 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 21356265-4 2011 In the present study, we used molecular dynamics simulation and autodocking studies to provide models for gliclazide-bound complexes of CYP2C9*2, *3 and *2/*3 mutants, which give insight into CYP2C9-gliclazide interactions and explain the reduced enzymatic activity seen in these variants. Gliclazide 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21356265-4 2011 In the present study, we used molecular dynamics simulation and autodocking studies to provide models for gliclazide-bound complexes of CYP2C9*2, *3 and *2/*3 mutants, which give insight into CYP2C9-gliclazide interactions and explain the reduced enzymatic activity seen in these variants. Gliclazide 199-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 21356265-4 2011 In the present study, we used molecular dynamics simulation and autodocking studies to provide models for gliclazide-bound complexes of CYP2C9*2, *3 and *2/*3 mutants, which give insight into CYP2C9-gliclazide interactions and explain the reduced enzymatic activity seen in these variants. Gliclazide 199-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21318593-2 2011 This study sought to systematically investigate the effects of genetic polymorphisms of VKORC1, STX4A, CYP2C9, CYP3A4, and GGCX and interacting drugs on the initial responses to warfarin in Chinese patients with heart valve replacement (HVR). Warfarin 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 21318593-8 2011 CONCLUSIONS: Not only VKORC1 and CYP2C9 genotypes, but also interacting drugs, had a significant impact on the variability of the initial response to warfarin. Warfarin 150-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 21451434-0 2011 Characterization of a novel CYP2C9 gene mutation and structural bioinformatic protein analysis in a warfarin hypersensitive patient. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 21349323-2 2011 The median inhibitory concentrations (IC50) of asiaticoside and madecassoside were determined for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4. asiaticoside 47-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 21349323-2 2011 The median inhibitory concentrations (IC50) of asiaticoside and madecassoside were determined for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4. madecassoside 64-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 21815482-4 2011 Recent studies have shown that genetic variations in a subunit of VKOR complex, VKORC1, and in cytochrome P 450 (CYP) 2C9 genes are strong determinants of individuals" warfarin sensitivity. Warfarin 168-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-121 21815482-5 2011 Algorithms have been proposed to predict warfarin doses, and about 55% of the variance in warfarin dose could be attributed to variations in the VKORC1 and CYP2C9 genes together with age, sex, body-surface area, and presence or absence of heart valve replacement. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 21815482-5 2011 Algorithms have been proposed to predict warfarin doses, and about 55% of the variance in warfarin dose could be attributed to variations in the VKORC1 and CYP2C9 genes together with age, sex, body-surface area, and presence or absence of heart valve replacement. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 21815482-6 2011 Contributions of polymorphisms in VKORC1 and CYP2C9 to inter-individual differences of warfarin dose, however, are different among races. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21451434-4 2011 We genotyped common polymorphisms in VKORC1, CYP2C9, and CYP4F2 genes, known to be involved in warfarin dosing. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21480951-1 2011 AIM: To investigate the impact of genetic polymorphisms in CYP2D6, CYP3A5, CYP2C9 and CYP2C19 on the pharmacokinetics of tamoxifen and its metabolites in Asian breast cancer patients. Tamoxifen 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 21497079-3 2011 The MWCNT structuring improves the sensitivity from 5.1 to 20.5 nA/mM mm(2) in case of CYP2B4-mediated Benzphetamine detection, from 0.26 to 0.63 nA/muM mm(2) in case of CYP3A4-mediated Cyclophosphamide detection, and from 0.11 to 0.25 nA/muM mm(2) in case of CYP2C9-mediated Naproxen detection. Benzphetamine 103-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 260-266 21289076-1 2011 According to published in vitro studies, cytochrome P450 3A4 catalyzes montelukast 21-hydroxylation (M5 formation), whereas CYP2C9 catalyzes 36-hydroxylation (M6), the primary step in the main metabolic pathway of montelukast. montelukast 214-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 21289076-6 2011 The CYP2C8 inhibitors gemfibrozil 1-O-beta glucuronide and trimethoprim inhibited the depletion of 0.02 muM montelukast and formation of M6 from 0.05 muM montelukast more potently than did the CYP2C9 inhibitor sulfaphenazole. Trimethoprim 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 21213107-2 2011 As the genetically polymorphic cytochrome P450 (CYP), enzyme CYP2C9 is mainly responsible for the hepatic metabolism of sulfonylureas (SUs), we hypothesized that the slow-metabolizer genotypes *2/*2, *2/*3, and *3/*3 might be overrepresented in type 2 diabetic patients with SH. Sulfonylurea Compounds 120-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21321060-0 2011 CYP2C9-mediated metabolic activation of losartan detected by a highly sensitive cell-based screening assay. Losartan 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21321060-6 2011 Metabolic activation of losartan by CYP2C9 has never been reported, although the metabolic activations of benzbromarone and tienilic acid have been reported. Losartan 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 21321060-8 2011 Two CYP2C9-specific semicarbazide adducts of losartan (S1 and S2) were detected. carbamylhydrazine 20-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21321060-8 2011 Two CYP2C9-specific semicarbazide adducts of losartan (S1 and S2) were detected. Losartan 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 21321060-10 2011 In conclusion, a highly sensitive cell-based assay to evaluate CYP2C9-mediated metabolic activation was established, and we found for the first time that CYP2C9 is involved in the metabolic activation of losartan. Losartan 204-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21321060-10 2011 In conclusion, a highly sensitive cell-based assay to evaluate CYP2C9-mediated metabolic activation was established, and we found for the first time that CYP2C9 is involved in the metabolic activation of losartan. Losartan 204-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 21220010-5 2011 The IC(50) concerning the CYP 2C9-mediated 4-hydroxylation of diclofenac has been calculated to be 0.04 mM (PS80), 0.30 mM (TPGS), 0.07 mM (sucrose laurate), 0.03 mM (CrEL), and 0.03 mM (Cr RH 40). Diclofenac 62-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-33 21220010-5 2011 The IC(50) concerning the CYP 2C9-mediated 4-hydroxylation of diclofenac has been calculated to be 0.04 mM (PS80), 0.30 mM (TPGS), 0.07 mM (sucrose laurate), 0.03 mM (CrEL), and 0.03 mM (Cr RH 40). sucrose monolaurate 140-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-33 21213107-2 2011 As the genetically polymorphic cytochrome P450 (CYP), enzyme CYP2C9 is mainly responsible for the hepatic metabolism of sulfonylureas (SUs), we hypothesized that the slow-metabolizer genotypes *2/*2, *2/*3, and *3/*3 might be overrepresented in type 2 diabetic patients with SH. Sulfonylurea Compounds 135-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21213107-9 2011 However, in the control group, patients with CYP2C9 genotypes, predicting slower metabolism of SU drugs, were treated with significantly lower doses (p = 0.027) than were extensive metabolizers, whereas in the patient group with severe hypoglycemia, the dose was the same for all genotype groups. Sulfonylurea Compounds 95-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21220010-1 2011 The purpose of the study was to investigate the impact of commonly used non-ionic surfactants on cytochrome P450 (CYP) 3A4-mediated metabolism of testosterone and the CYP2C9-mediated metabolism of diclofenac. Diclofenac 197-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 21543033-1 2011 OBJECTIVE: To investigate the effect of the estimated highest therapeutic dose of linagliptin (5 mg) on the pharmacokinetics and pharmacodynamics of warfarin, a CYP2C9 substrate. Linagliptin 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 21543033-1 2011 OBJECTIVE: To investigate the effect of the estimated highest therapeutic dose of linagliptin (5 mg) on the pharmacokinetics and pharmacodynamics of warfarin, a CYP2C9 substrate. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Fluvoxamine 0-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 21292004-6 2011 NCOA6 enhances the synergistic activation of CYP2C9 and CYP3A4 promoter activity by PXR and HNF4alpha in the presence of rifampicin. Rifampin 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21294626-4 2011 CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were identified as the major enzymes responsible for the formation of the two O-desmethyl magnolins (M1 and M2), on the basis of a combination of correlation analysis and experiments, including immunoinhibition of magnolin in human liver microsomes and metabolism of magnolin by human cDNA-expressed CYP enzymes. magnolin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 21294626-4 2011 CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were identified as the major enzymes responsible for the formation of the two O-desmethyl magnolins (M1 and M2), on the basis of a combination of correlation analysis and experiments, including immunoinhibition of magnolin in human liver microsomes and metabolism of magnolin by human cDNA-expressed CYP enzymes. magnolin 250-258 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 21277363-4 2011 Fluvoxamine maleate, gemfibrozil, amiodarone hydrochloride, omeprazole, quinidine, diethyldithiocarbamic acid and ketoconazole were successfully applied as test inhibitors for CYP1A2, CYP2C8, CYP2C9, CYP2C19*1, CYP2D6*1, CYP2E1 and CYP3A4/5 in HLMs, respectively. Ketoconazole 114-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 20927605-7 2011 In addition, the preparative of synthesis of 2.8 g of 4"-hydroxydiclofenac was achieved by whole-cell biotransformation of diclofenac using a CPR-CYP2C9 coexpressing fission yeast strain. 4'-hydroxydiclofenac 54-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 20927605-7 2011 In addition, the preparative of synthesis of 2.8 g of 4"-hydroxydiclofenac was achieved by whole-cell biotransformation of diclofenac using a CPR-CYP2C9 coexpressing fission yeast strain. Diclofenac 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 21395648-6 2011 The oral clearance of meloxicam was significantly lower in the CYP2C9*1/*13 group (37.9% of wild type) than in the CYP2C9*1/*1 group. Meloxicam 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 21395648-7 2011 The t(1/2) of meloxicam was 1.84-fold longer in the CYP2C9*1/*13 group than in the CYP2C9*1/*1 group. Meloxicam 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 21395648-0 2011 Effects of CYP2C9*1/*13 on the pharmacokinetics and pharmacodynamics of meloxicam. Meloxicam 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 21395648-7 2011 The t(1/2) of meloxicam was 1.84-fold longer in the CYP2C9*1/*13 group than in the CYP2C9*1/*1 group. Meloxicam 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 21395648-1 2011 AIMS: To determine the effects of the CYP2C9*1/*13 genotype on the pharmacokinetics and pharmacodynamics of meloxicam in Korean subjects. Meloxicam 108-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 21395648-9 2011 CONCLUSIONS: The CYP2C9*1/*13 genotype is associated with decreased metabolism and increased pharmacodynamic effects of meloxicam. Meloxicam 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 21395648-2 2011 METHODS: Meloxicam (15 mg) was orally administered to 21 healthy Korean volunteers with either the CYP2C9*1/*1 or the CYP2C9*1/*13 genotype. Meloxicam 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 21395648-2 2011 METHODS: Meloxicam (15 mg) was orally administered to 21 healthy Korean volunteers with either the CYP2C9*1/*1 or the CYP2C9*1/*13 genotype. Meloxicam 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 21177984-5 2011 Ticagrelor moderately inhibited CYP2C9 activity in human liver microsomes with an IC(50) of 10.5 muM, while exhibiting little or no inhibition of CYP1A2, CYP2B6, CYP2C8, CYP2C19, CYP2D6, and CYP2E1. Ticagrelor 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 21395648-5 2011 RESULTS: The AUC(0, ) and C(max) of meloxicam were 2.43- and 1.46-fold higher in the CYP2C9*1/*13 group than in the CYP2C9*1/*1 group, respectively. Meloxicam 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21395648-5 2011 RESULTS: The AUC(0, ) and C(max) of meloxicam were 2.43- and 1.46-fold higher in the CYP2C9*1/*13 group than in the CYP2C9*1/*1 group, respectively. Meloxicam 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 21395648-6 2011 The oral clearance of meloxicam was significantly lower in the CYP2C9*1/*13 group (37.9% of wild type) than in the CYP2C9*1/*1 group. Meloxicam 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21768671-0 2011 Association and treatment response to capecitabine-based chemoradiotherapy with CYP2C9 polymorphism in head and neck cancer. Capecitabine 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 21537551-0 2011 CYP2C9 polymorphism in patients with epilepsy: genotypic frequency analyzes and phenytoin adverse reactions correlation. Phenytoin 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21537551-12 2011 The role of CYP2C9 polymorphism influence on phenytoin adverse reaction remains to be determined since some literature evidence and our data found negative results. Phenytoin 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 21270790-0 2011 The missing association: sequencing-based discovery of novel SNPs in VKORC1 and CYP2C9 that affect warfarin dose in African Americans. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 20458343-4 2011 The CYP2C9*2 frequency was expected to be lower in the indigenous MT than in the two other groups, and lower in MM than in SP as in our earlier study. sp 123-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 20458343-8 2011 The frequencies of CYP2C9*2 were 0.01, 0.07, 0.08, and 0.16 for MT, MM, MA, and SP subjects, respectively. sp 80-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 20458343-9 2011 In agreement with our hypothesis, CYP2C9*2 was significantly lower in the Mexican populations than in the SP (P<0.05), and among Mexicans in the MT than in the MM and MA groups (P<0.05), which presented similar frequencies. sp 106-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20458343-10 2011 Moreover, the frequency of CYP2C9*3 was found to be lower (P<0.05) in MM (0.015) and MT (0.015) than in MA (0.06) and SP (0.08). sp 121-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 21483991-8 2011 The group that required low dose warfarin (n=25) showed significantly higher CYP2C9 polymorphism, required 40% less warfarin and had a higher rate of bleeding (5% versus 1.8%). Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 21483991-10 2011 Patients with the CYP2C9 polymorphism required 40% less warfarin and had more serious bleeds. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 21270790-5 2011 Through ethnicity-specific warfarin dose model building in 330 African Americans, we identified two novel genetic associations with higher warfarin dose, namely, VKORC1-8191 (rs61162043, P = 0.0041) and 18786 in CYP2C9 (rs7089580, P = 0.035). Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 21326313-6 2011 The SNPs of VKORC1, CYP2C9, CYP4F2 and GGCX showed significant correlation with warfarin dose. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 21326313-10 2011 This study shows that age, gender, BSA, INR and VKORC1, CYP2C9 and CYP4F2 polymorphism affect warfarin dose requirements in Koreans. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 21294704-7 2011 It has also shown mild activation effect on CYP 269 and CYP 2C9 enzymes, indicating the usefulness of thiosemicarbazones as anticonvulsants. Thiosemicarbazones 102-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-63 21270790-1 2011 It is well recognized that the genetic variants VKORC1-1639, CYP2C9*2, and CYP2C9*3 contribute to warfarin dose response. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21270790-1 2011 It is well recognized that the genetic variants VKORC1-1639, CYP2C9*2, and CYP2C9*3 contribute to warfarin dose response. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 21270790-5 2011 Through ethnicity-specific warfarin dose model building in 330 African Americans, we identified two novel genetic associations with higher warfarin dose, namely, VKORC1-8191 (rs61162043, P = 0.0041) and 18786 in CYP2C9 (rs7089580, P = 0.035). Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 21185752-8 2011 The combination of the VKORC1-1639G>A and CYP2C9 genotypes and clinical factors explained 56% of the inter-patient variability in warfarin dose. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21336516-7 2011 The most selective inhibitors available are furafylline (in co-incubation and pre-incubation conditions) for CYP1A2, 2-phenyl-2-(1-piperidinyl)propane (PPP) for CYP2B6, montelukast for CYP2C8, sulfaphenazole for CYP2C9, (-)-N-3-benzyl-phenobarbital for CYP2C19 and quinidine for CYP2D6. 2-phenyl-2-(1-piperidinyl)propane 152-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 21553654-4 2011 In vitro inhibitory effects of DES on CYP isoforms were investigated, and the results showed that DES could competitively inhibit CYP3A4, CYP2C8, CYP2C9 and CYP2E1. Diethylstilbestrol 31-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 21553654-4 2011 In vitro inhibitory effects of DES on CYP isoforms were investigated, and the results showed that DES could competitively inhibit CYP3A4, CYP2C8, CYP2C9 and CYP2E1. Diethylstilbestrol 98-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 21553654-6 2011 Based on peak serum DES level after drip influsion of 500 mg of fosfestrol (DES diphosphate) in patients, [I]/Ki was calculated to be 4.3, 6.2, 3.7 and 2.3 for CYP3A4, CYP2C9, CYP2E1 and CYP2C8, which suggested that DES was likely to induce in vivo DDI through inhibition of these four major CYP isoforms. fosfestrol 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 21553655-1 2011 Polymorphisms in cytochrome P450 (CYP) 2C9 and the vitamin K oxide reductase complex subunit 1 (VKORC1) greatly affect the maintenance dose of warfarin. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-42 21194699-3 2011 Acetaminophen (CYP1A2), dextrorphan (CYP2D6), hydroxy-chlorzoxazone (CYP2E1) and hydroxy-mephenytoin (CYP2C19) can be derivatized because of the presence of a phenolic OH, whereas hydroxy-midazolam (CYP3A4) and hydroxy-tolbutamide (CYP2C9) remain unchanged. hydroxy-chlorzoxazone 46-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 21194699-3 2011 Acetaminophen (CYP1A2), dextrorphan (CYP2D6), hydroxy-chlorzoxazone (CYP2E1) and hydroxy-mephenytoin (CYP2C19) can be derivatized because of the presence of a phenolic OH, whereas hydroxy-midazolam (CYP3A4) and hydroxy-tolbutamide (CYP2C9) remain unchanged. hydroxy-mephenytoin 81-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 21194699-3 2011 Acetaminophen (CYP1A2), dextrorphan (CYP2D6), hydroxy-chlorzoxazone (CYP2E1) and hydroxy-mephenytoin (CYP2C19) can be derivatized because of the presence of a phenolic OH, whereas hydroxy-midazolam (CYP3A4) and hydroxy-tolbutamide (CYP2C9) remain unchanged. hydroxy-midazolam 180-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 21194699-3 2011 Acetaminophen (CYP1A2), dextrorphan (CYP2D6), hydroxy-chlorzoxazone (CYP2E1) and hydroxy-mephenytoin (CYP2C19) can be derivatized because of the presence of a phenolic OH, whereas hydroxy-midazolam (CYP3A4) and hydroxy-tolbutamide (CYP2C9) remain unchanged. hydroxymethyltolbutamide 211-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 21194699-3 2011 Acetaminophen (CYP1A2), dextrorphan (CYP2D6), hydroxy-chlorzoxazone (CYP2E1) and hydroxy-mephenytoin (CYP2C19) can be derivatized because of the presence of a phenolic OH, whereas hydroxy-midazolam (CYP3A4) and hydroxy-tolbutamide (CYP2C9) remain unchanged. Acetaminophen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 21223634-8 2011 However, one subject"s data was suggestive of being poor metabolizer of flurbiprofen which supports the presence of CYP2C9 polymorphism contributing to inter-individual differences in flurbiprofen disposition. Flurbiprofen 72-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 20931329-2 2011 Like montelukast, zafirlukast is a substrate of CYP2C9 and CYP3A4 and a potent inhibitor of CYP2C8 in vitro. montelukast 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 20931329-2 2011 Like montelukast, zafirlukast is a substrate of CYP2C9 and CYP3A4 and a potent inhibitor of CYP2C8 in vitro. zafirlukast 18-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 21223634-8 2011 However, one subject"s data was suggestive of being poor metabolizer of flurbiprofen which supports the presence of CYP2C9 polymorphism contributing to inter-individual differences in flurbiprofen disposition. Flurbiprofen 184-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 20641061-5 2011 Among seven major CYP isoforms tested, corynoline showed strong inhibitory effects on the activities of CYP3A4 and CYP2C9, with an IC(50) of 3.3 +- 0.9 microm and 31.5 +- 0.5 microm, respectively. corynoline 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 21469766-1 2011 BACKGROUND: Variant alleles of vitamin K epoxide reductase complex subunit 1 gene (VKORC1), the target molecule of vitamin K antagonists, and of cytochrome P450 (CYP) 2C9, an enzyme involved in coumarin metabolism, affect the anticoagulant response of coumarins, which have a narrow therapeutic window. coumarin 194-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-170 21469766-1 2011 BACKGROUND: Variant alleles of vitamin K epoxide reductase complex subunit 1 gene (VKORC1), the target molecule of vitamin K antagonists, and of cytochrome P450 (CYP) 2C9, an enzyme involved in coumarin metabolism, affect the anticoagulant response of coumarins, which have a narrow therapeutic window. Coumarins 252-261 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-170 20641061-6 2011 Kinetic analysis showed that inhibition of CYP3A4 by corynoline was best fit to a noncompetitive manner with K(i) of 3.2 microm, while inhibition of CYP2C9 by corynoline was best fit to a competitive manner with K(i) of 6.3 microm. corynoline 159-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 21167292-7 2011 Selective inhibition of CYP2C9 decreased tumor cell proliferation (KYSE30, PT1590 and OE19) in vitro, which was abolished by 11,12-epoxyeicosatrienoic acid (11,12-EET). pt1590 75-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 21167292-7 2011 Selective inhibition of CYP2C9 decreased tumor cell proliferation (KYSE30, PT1590 and OE19) in vitro, which was abolished by 11,12-epoxyeicosatrienoic acid (11,12-EET). 11,12-epoxy-5,8,14-eicosatrienoic acid 125-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 22540071-8 2011 CONCLUSIONS: sulfamethoxazole is catalysed by CYP2C9 and/or myeloperoxidase. Sulfamethoxazole 13-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 21167292-7 2011 Selective inhibition of CYP2C9 decreased tumor cell proliferation (KYSE30, PT1590 and OE19) in vitro, which was abolished by 11,12-epoxyeicosatrienoic acid (11,12-EET). 11,12-epoxy-5,8,14-eicosatrienoic acid 157-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 21691466-4 2011 We sought in a pilot study to identify if testing for common CYP2C9 and VKORC1 single nucleotide polymorphisms (SNPs) in patients who were likely to begin warfarin treatment was feasible in an ED setting. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21157402-2 2011 (S)-Warfarin has more potent anticoagulant activity than (R)-warfarin and is metabolized mainly to (S)-7-hydroxywarfarin by CYP2C9. Warfarin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 21157402-2 2011 (S)-Warfarin has more potent anticoagulant activity than (R)-warfarin and is metabolized mainly to (S)-7-hydroxywarfarin by CYP2C9. 7-hydroxywarfarin 99-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 21532165-4 2011 Recombinant human CYP2C9 exhibited appreciable catalytic activity with respect to 6-HNA formation from 6-MNA. 6-Mna 103-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 21532165-6 2011 Sulfaphenazole (a selective inhibitor of CYP2C9) inhibited the formation of 6-HNA in pooled human microsomes by 89%, but failed to inhibit this reaction in rat liver microsomes. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 21142266-3 2011 In vivo, etravirine is a substrate for, and weak inducer of, the hepatic cytochrome P450 (CYP) isoenzyme 3A4 and a substrate and weak inhibitor of CYP2C9 and CYP2C19. etravirine 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 21184622-2 2011 Nilotinib has shown competitive inhibition of cytochrome P450 enzyme (CYP) 2C9 in vitro, but its effect on CYP2C9 activity in humans is unknown. nilotinib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-78 21184622-2 2011 Nilotinib has shown competitive inhibition of cytochrome P450 enzyme (CYP) 2C9 in vitro, but its effect on CYP2C9 activity in humans is unknown. nilotinib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 21184622-3 2011 This study evaluated the effects of nilotinib on the pharmacokinetics and pharmacodynamics of warfarin, a sensitive CYP2C9 substrate, in healthy subjects. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 21084763-1 2011 The aim of this study was to investigate the effect of the dipeptidyl peptidase-4 inhibitor linagliptin on the pharmacokinetics of glyburide (a CYP2C9 and CYP3A4 substrate) and vice versa. Linagliptin 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Phenytoin 65-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Warfarin 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Tolbutamide 88-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Glipizide 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Diclofenac 112-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-1 2011 CYP2C9 metabolizes more than 100 clinically used drugs including phenytoin, S-warfarin, tolbutamide, glipizide, diclofenac, and losartan with varying contributions. Losartan 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21182487-10 2011 Polymorphisms in CYP2C9 have the potential to affect the clearance and clinical response of CYP2C9 substrate drugs with low therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Warfarin 152-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 21182487-10 2011 Polymorphisms in CYP2C9 have the potential to affect the clearance and clinical response of CYP2C9 substrate drugs with low therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Warfarin 152-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 21182487-10 2011 Polymorphisms in CYP2C9 have the potential to affect the clearance and clinical response of CYP2C9 substrate drugs with low therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Phenytoin 162-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 21182487-10 2011 Polymorphisms in CYP2C9 have the potential to affect the clearance and clinical response of CYP2C9 substrate drugs with low therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Phenytoin 162-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 21557672-5 2011 In light of the present evidence, patients treated with PHT could benefit from CYP2C9 and CYP2C19 genotyping/phenotyping. Phenytoin 56-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 21084763-1 2011 The aim of this study was to investigate the effect of the dipeptidyl peptidase-4 inhibitor linagliptin on the pharmacokinetics of glyburide (a CYP2C9 and CYP3A4 substrate) and vice versa. Glyburide 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 21084764-5 2011 In patients harboring VKORC1 diplotypes associated with low warfarin requirements, a linear regression model which included age, weight, CYP2C9 and CYP4F2 variants accounted for 38% of the variability in warfarin dose. Warfarin 204-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21176721-6 2011 The multiple linear regression model for warfarin dose indicated significant contributions from the VKORC1 (1173C > T) genotype (r2 = 0.355; p < 0.001), and age, body weight, and CYP2C9 and VKORC1 genotype together (r2 = 0.513; p < 0.001). Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 21127708-5 2011 Our results indicate that pharmacogenetic testing for VKORC1*2, CYP2C9*2 and CYP2C9*3 is more informative regarding warfarin dose requirements than testing for VKORC1*3, VKORC1*4, and CYP4F2 (1347 C > T) polymorphisms. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 21321468-0 2011 Clinical significance of combined CYP2C9 and VKORC1 genotypes in Japanese patients requiring warfarin. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 21321468-9 2011 Thus, CYP2C9 and VKORC1 haplotype analysis allows prediction of warfarin maintenance dosage. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 20852447-0 2011 Role of CYP2C9 genetic variants for salt sensitivity and the regulation of the renin-angiotensin-aldosterone system in normotensive men. Salts 36-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 20852447-1 2011 OBJECTIVE: Cytochrome P450 (CYP) 2C9 gene polymorphisms have been implicated in regulation of the renin-angiotensin-aldosterone system (RAAS) and salt sensitivity in hypertensive patients. Salts 146-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-36 20852447-6 2011 CYP2C9*3 carriers showed a significantly lower PRA compared with CYP2C9*1/*1 individuals in the overall study cohort (high salt: 0.39 +- 0.05 vs. 0.62 +- 0.04 ng/ml per h, P = 0.009; low salt: 2.19 +- 0.27 vs. 2.87 +- 0.13 ng/ml per h, P = 0.013). Salts 123-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20852447-6 2011 CYP2C9*3 carriers showed a significantly lower PRA compared with CYP2C9*1/*1 individuals in the overall study cohort (high salt: 0.39 +- 0.05 vs. 0.62 +- 0.04 ng/ml per h, P = 0.009; low salt: 2.19 +- 0.27 vs. 2.87 +- 0.13 ng/ml per h, P = 0.013). Salts 187-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20852447-7 2011 Salt-sensitive CYP2C9*3 carriers exhibited the lowest PRA values and significantly lower 24 h sodium excretion rates during high-salt intake (P = 0.005 vs. CYP2C9*1/*1). Salts 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20852447-7 2011 Salt-sensitive CYP2C9*3 carriers exhibited the lowest PRA values and significantly lower 24 h sodium excretion rates during high-salt intake (P = 0.005 vs. CYP2C9*1/*1). Salts 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 20852447-7 2011 Salt-sensitive CYP2C9*3 carriers exhibited the lowest PRA values and significantly lower 24 h sodium excretion rates during high-salt intake (P = 0.005 vs. CYP2C9*1/*1). Sodium 94-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20852447-7 2011 Salt-sensitive CYP2C9*3 carriers exhibited the lowest PRA values and significantly lower 24 h sodium excretion rates during high-salt intake (P = 0.005 vs. CYP2C9*1/*1). Salts 129-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20852447-8 2011 Lower plasma aldosterone concentrations were only observed in salt-resistant CYP2C9*3 carriers under low salt (P = 0.039). Salts 62-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 20852447-8 2011 Lower plasma aldosterone concentrations were only observed in salt-resistant CYP2C9*3 carriers under low salt (P = 0.039). Salts 105-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 20852447-11 2011 Further studies are needed to analyze the long-term effects of CYP2C9*3 for salt sensitivity and hypertensive diseases. Salts 76-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 21160360-0 2011 CYP2C9 variants and blood pressure response to salt: when salt sensitivity meets pharmacogenomics. Salts 58-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21127708-5 2011 Our results indicate that pharmacogenetic testing for VKORC1*2, CYP2C9*2 and CYP2C9*3 is more informative regarding warfarin dose requirements than testing for VKORC1*3, VKORC1*4, and CYP4F2 (1347 C > T) polymorphisms. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 20585834-9 2011 CYP2C9 and VKORC1 genotype, age, weight and vitamin K intake were identified by stepwise regression modelling to produce the best model for estimating warfarin dose (R (2) = 68%, P < 0.001). Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21681008-1 2011 UNLABELLED: A clinical effect of warfarin depends on highly polymorphic drug-metabolizing (CYP2C9) and drug-target (VKORC1) enzymes. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 20585834-1 2011 CYP2C9 and VKORC1 genotypes could be used to predict warfarin requirement. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 21681008-2 2011 The objective of this study was to investigate the impact of CYP2C9*2, CYP2C9*3, and VKORC1 (G-1639A) polymorphisms on the variability of warfarin dosage requirements in Lithuanian patients after heart valve replacement. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 21681008-2 2011 The objective of this study was to investigate the impact of CYP2C9*2, CYP2C9*3, and VKORC1 (G-1639A) polymorphisms on the variability of warfarin dosage requirements in Lithuanian patients after heart valve replacement. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 21681008-8 2011 WT CYP2C9*1/*1 genotype was associated with a higher daily warfarin dosage (5.84 mg [SD, 2.84]) as compared to other CYP2C9 genotypes. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 21681008-10 2011 Patients having CYP2C9*1/*1 or 1/*2 in combination with VKORC1 (G/G) or (G/A) genotypes required the highest daily warfarin dosage in comparison to other combinations of genotypes. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 21681008-12 2011 Analysis of combined CYP2C9 and VKORC1 gene variants allows the prediction of warfarin dosage. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 23226058-5 2011 This is the case with warfarin, with lower doses often necessary in carriers of a variant CYP2C9*2 or *3 allele to avoid supratherapeutic anticoagulation. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 21063236-1 2011 BACKGROUND: Genome-wide association studies (GWAS) on warfarin and acenocoumarol showed that interindividual dosage variation is mainly associated with single nucleotide polymorphisms (SNPs) in VKORC1 and to a lesser extent in CYP2C9 and CYP4F2. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 227-233 21063236-1 2011 BACKGROUND: Genome-wide association studies (GWAS) on warfarin and acenocoumarol showed that interindividual dosage variation is mainly associated with single nucleotide polymorphisms (SNPs) in VKORC1 and to a lesser extent in CYP2C9 and CYP4F2. Acenocoumarol 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 227-233 21063236-6 2011 RESULTS: Within our study population (N=244), VKORC1, CYP2C9, CYP4F2 genotypes together explained 46% of phenprocoumon maintenance dosage variation. Phenprocoumon 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 21174619-0 2011 VKORC1, CYP2C9 and CYP4F2 genetic-based algorithm for warfarin dosing: an Italian retrospective study. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 21142408-2 2011 Both clopidogrel and the oral hypoglycemic drug class sulfonylureas are metabolized by the iso-enzyme CYP2C9. Clopidogrel 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 21462897-6 2011 At concentration up to 50 micromol x L(-1), GA inhibited CYP2C19, CYP2C9 and CYP3A4 enzyme activities with the inhibitory potencies up to 50%. Glycyrrhetinic Acid 44-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 21142408-2 2011 Both clopidogrel and the oral hypoglycemic drug class sulfonylureas are metabolized by the iso-enzyme CYP2C9. Sulfonylurea Compounds 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 22114699-5 2011 Regression analysis demonstrated that CYP2C9 genotype, kidney function, and gender were independent determinants of S-warfarin clearance. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 21786578-1 2011 AIM: To investigate frequency of carriage of genetic polymorphisms CYP2C9 and VKORC1 in patients with venous thromboembolic complications (VTEC) in Moscow population given warfarin treatment and effects of this carriage on stability of anticoagulation and frequency of hemorrhagic complications (HC) in warfarin treatment. Warfarin 172-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 21786578-1 2011 AIM: To investigate frequency of carriage of genetic polymorphisms CYP2C9 and VKORC1 in patients with venous thromboembolic complications (VTEC) in Moscow population given warfarin treatment and effects of this carriage on stability of anticoagulation and frequency of hemorrhagic complications (HC) in warfarin treatment. Warfarin 303-311 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 21786578-10 2011 A mean warfarin dose, supporting an adequaite INR, was asspciated with both genotype CYP2C9 and VKORC1. Warfarin 7-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 21786578-11 2011 Warfarin doses were highest in carriers of wile genotypes CYP2C9 and VKORC1 (6,9 and 8,8 mg/day), the lowest--in patients with genotypes CYP2C9*1/*3 and VKORC1 (4,5 and 4,0 mg/day). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 21786578-11 2011 Warfarin doses were highest in carriers of wile genotypes CYP2C9 and VKORC1 (6,9 and 8,8 mg/day), the lowest--in patients with genotypes CYP2C9*1/*3 and VKORC1 (4,5 and 4,0 mg/day). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21786578-15 2011 The results of multifactorial regression analysis also indicated that carriage of genotype CYP2C9*1/*3, a female gender and the range of INR in warfarin treatment > or = 2,66 are independent predictors of HC in VTEC patients on warfarin treatment. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 21786578-15 2011 The results of multifactorial regression analysis also indicated that carriage of genotype CYP2C9*1/*3, a female gender and the range of INR in warfarin treatment > or = 2,66 are independent predictors of HC in VTEC patients on warfarin treatment. Warfarin 231-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 21786578-16 2011 CONCLUSION: Carriage of gene CYP2C9 and VKORC1 polymorphisms affects suppoting dose of warfarin and rate of hemorrhage in patients with VTEC in Moscow population. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 21786578-17 2011 Frequency of HC is the highest in carriers of genotypes CYP2C9*1/*3 and AA VKORC1, they need minimal supporting dose of warfarin. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 21786578-18 2011 Carriage of genotype CYP2C9*1/*3 in line with a female gender and instability of INR is an independent predictor of HC in VTEC patients in Moscow population on warfarin treatment. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 21532277-6 2011 These findings indicate that polymorphism of CYP2C9 and CYP2C19 plays an important role in phenytoin metabolism in children. Phenytoin 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 21532277-7 2011 With a total processing time for this assay of less than 3 hours, prediction of the optimal phenytoin dosage based on the CYP2C9 and CYP2C19 genotypes will be possible before commencement of therapy, resulting in the prevention of phenytoin overdoses in pediatric patients with epilepsy. Phenytoin 92-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 21532277-7 2011 With a total processing time for this assay of less than 3 hours, prediction of the optimal phenytoin dosage based on the CYP2C9 and CYP2C19 genotypes will be possible before commencement of therapy, resulting in the prevention of phenytoin overdoses in pediatric patients with epilepsy. Phenytoin 231-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 20609441-5 2010 Norketamine was formed by CYP3A4, CYP2C19, CYP2B6, CYP2A6, CYP2D6 and CYP2C9, whereas CYP2B6 and CYP2A6 were identified to be the only enzymes which enable the hydroxylation of norketamine. norketamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 23120787-2 2011 The development of undesirable pharmacological phenomena at reception of average therapeutic doses of valproic acid has been caused by the primary (idiopathic) and secondary (valproate-induced) infringement of a folic cycle against a combination of CYP2C9*3 gene polymorphism and a mutation in the MTHFR gene. Valproic Acid 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 23120787-2 2011 The development of undesirable pharmacological phenomena at reception of average therapeutic doses of valproic acid has been caused by the primary (idiopathic) and secondary (valproate-induced) infringement of a folic cycle against a combination of CYP2C9*3 gene polymorphism and a mutation in the MTHFR gene. Valproic Acid 175-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Phenytoin 126-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Phenytoin 126-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Warfarin 137-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Warfarin 137-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Tolbutamide 149-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Tolbutamide 149-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Losartan 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19715737-1 2010 Human cytochrome P450 2C9 (CYP2C9) accounts for ~20% of hepatic total CYP content and metabolizes ~15% clinical drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and many nonsteroidal anti-inflammatory agents (NSAIDs). Losartan 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 20736323-6 2010 The inhibitory potency of DBZ toward CYP3A4 was greater than that toward other P450 isoforms, including CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6. dibenzazepine 26-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 21193152-7 2010 Pitavastatin is only slightly metabolised by cytochrome P450 (CYP) 2C9 and not at all by CYP3A4. pitavastatin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-70 20851877-4 2010 Based on the kinetic data and average contents of the P450 isoforms in the human liver, the putative contribution of P450s for sesamin metabolism was large in the order of CYP2C9, 1A2, 2C19, and 2D6. sesamin 127-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 20833655-8 2010 Our study also implied that common SNPs other than those in the CYP2C9, VKORC1 and CYP4F2 genes that show strong effect on the therapeutic warfarin dose might not exist. Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 40-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 40-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 237-244 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 20851877-5 2010 A good correlation was observed between sesamin catecholization activity and CYP2C9-specific activity in in vitro studies using 10 individual human liver microsomes, strongly suggesting that CYP2C9 is the most important P450 isoform for sesamin catecholization in human liver. sesamin 237-244 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 20851877-7 2010 We also examined the inhibitory effect of sesamin for P450 isoform-specific activities and found a mechanism-based inhibition of CYP2C9 by sesamin. sesamin 42-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 20851877-7 2010 We also examined the inhibitory effect of sesamin for P450 isoform-specific activities and found a mechanism-based inhibition of CYP2C9 by sesamin. sesamin 139-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 20851877-9 2010 Our findings strongly suggest that further studies are needed to reveal the interaction between sesamin and therapeutic drugs mainly metabolized by CYP2C9. sesamin 96-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 21261182-10 2010 We conclude that the rapid rise in INR observed upon the initiation of warfarin therapy and the final effective warfarin dose of 1.5 mg/day, are attributable in some part to the presence of two minor alleles in CYP2C9, which together significantly reduce warfarin metabolism. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 20578979-0 2010 Inhibitory effects of phthalimide derivatives on the activity of the hepatic cytochrome P450 monooxygenases CYP2C9 and CYP2C19. phthalimide 22-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 20578979-5 2010 In addition similar patterns of phthalimide inhibition for CYP2C9 and CYP2C19 were found, whereas the unrelated isoforms CYP2D6 and CYP3A4 were not specifically affected. phthalimide 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 20578979-7 2010 Analyses of structure-function relationships revealed that the substituent at the nitrogen atom in the isoindole ring is of crucial impact for the activity of CYP2C9/19. Nitrogen 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 20578979-7 2010 Analyses of structure-function relationships revealed that the substituent at the nitrogen atom in the isoindole ring is of crucial impact for the activity of CYP2C9/19. Isoindoles 103-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 20735727-10 2010 Consistently, R-warfarin significantly induced CYP3A4 and CYP2C9 mRNAs in cultures of primary human hepatocytes or in LS174T intestinal cells. Warfarin 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 20735727-12 2010 In addition, we showed that racemic 10- and 4"-hydroxywarfarins are also highly potent PXR ligands and inducers of CYP3A4 and CYP2C9 mRNA in human hepatocytes. 10- and 4"-hydroxywarfarins 36-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 20735727-13 2010 CONCLUSION: We showed that R-warfarin can significantly up-regulate major drug-metabolizing enzymes CYP3A4 and CYP2C9 in the liver and thus may cause drug-drug interactions (DDI) with co-administered drugs. Warfarin 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 21261182-10 2010 We conclude that the rapid rise in INR observed upon the initiation of warfarin therapy and the final effective warfarin dose of 1.5 mg/day, are attributable in some part to the presence of two minor alleles in CYP2C9, which together significantly reduce warfarin metabolism. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 21261182-10 2010 We conclude that the rapid rise in INR observed upon the initiation of warfarin therapy and the final effective warfarin dose of 1.5 mg/day, are attributable in some part to the presence of two minor alleles in CYP2C9, which together significantly reduce warfarin metabolism. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 21068649-1 2010 The objective of this study was to study the effect of CYP2C9 genetic polymorphism and undernourishment on free phenytoin concentrations in epileptic patients. Phenytoin 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 21068649-9 2010 The percentage increase in free phenytoin concentration by undernourishment, CYP2C9 allelic variants, and undernourishment cum CYP2C9 allelic variants were 127%, 290%, and 472%, respectively, compared with well-nourished patients with the wild-type CYP2C9 genotype (G1) group. Phenytoin 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 21068649-10 2010 The contribution of undernourishment and genetic factors (CYP2C9 allelic variant) for developing phenytoin toxicity was calculated to have an odds ratio of 37.3 (P < 0.0001). Phenytoin 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 21068649-11 2010 Undernourishment and variant CYP2C9 alleles elevate free phenytoin concentrations individually and in combination show additive effects. Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 20809476-7 2010 Fluvastatin and nifedipine increased the systemic exposure of nateglinide in rabbits, probably due to their inhibitory action on the metabolism of nateglinide by CYP2C5 (human CYP2C9). Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 21428138-7 2010 We analyzed the prevalence of the different CYP2C9 and VKORC1 genotypes in this population and found that the warfarin doses required to maintain patients at their desired anticoagulation target were significantly different among carriers of the different genotypes. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 20809476-7 2010 Fluvastatin and nifedipine increased the systemic exposure of nateglinide in rabbits, probably due to their inhibitory action on the metabolism of nateglinide by CYP2C5 (human CYP2C9). Nifedipine 16-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 20809476-7 2010 Fluvastatin and nifedipine increased the systemic exposure of nateglinide in rabbits, probably due to their inhibitory action on the metabolism of nateglinide by CYP2C5 (human CYP2C9). Nateglinide 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 20809476-7 2010 Fluvastatin and nifedipine increased the systemic exposure of nateglinide in rabbits, probably due to their inhibitory action on the metabolism of nateglinide by CYP2C5 (human CYP2C9). Nateglinide 147-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 21121772-0 2010 Effect of CYP2C9 polymorphisms on prescribed dose and time-to-stable dose of sulfonylureas in primary care patients with Type 2 diabetes mellitus. Sulfonylurea Compounds 77-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 20955109-5 2010 WHAT THE READER WILL GAIN: Following oral administration, idebenone is rapidly metabolized via oxidative shortening by a number CYP isoenzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4) to yield QS10, QS8, QS6 and QS4. idebenone 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 21121772-1 2010 AIMS: Sulfonylureas are mainly metabolized by the enzyme CYP2C9. Sulfonylurea Compounds 6-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 21121772-2 2010 Two allelic variants, CYP2C9*2 and CYP2C9*3, result in decreased metabolic capacity and have been associated with elevated sulfonylurea serum levels. Sulfonylurea Compounds 123-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 21031626-3 2010 The inhibition of seven CYP isoforms (CYP3A4, CYP1A2, CYP2A6, CYP2D6, CYP2C9, CYP2C8 and CYP2E1) by tiliroside was investigated using in vitro human liver microsomal incubation assays. tiliroside 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 21121772-2 2010 Two allelic variants, CYP2C9*2 and CYP2C9*3, result in decreased metabolic capacity and have been associated with elevated sulfonylurea serum levels. Sulfonylurea Compounds 123-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 21121772-4 2010 In this study, the effect of CYP2C9*2 and CYP2C9*3 alleles on prescribed dose and time-to-stable dose of sulfonylureas was investigated. Sulfonylurea Compounds 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 21031626-4 2010 The results showed that tiliroside strongly inhibited the activity of CYP3A4 (IC(50) = 9.0 +- 1.7 mum), CYP2C8 (IC(50) = 12.1 +- 0.9 mum) and CYP2C9 (IC(50) = 10.2 +- 0.9 mum) with other CYP isoforms negligibly influenced. tiliroside 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 21121772-4 2010 In this study, the effect of CYP2C9*2 and CYP2C9*3 alleles on prescribed dose and time-to-stable dose of sulfonylureas was investigated. Sulfonylurea Compounds 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 21031626-8 2010 Therefore, attention should be given to the probable drug-drug interaction between tiliroside-containing herbs and substrates of CYP3A4, CYP2C9 and CYP2C8. tiliroside 83-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 21121772-8 2010 However, a trend towards a lower stable glimepiride dose for carriers of the CYP2C9*3 allele was observed. glimepiride 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 21044367-9 2010 We find that our method can increase the power of the GWAS to identify both VKORC1 and CYP2C9 as warfarin pharmacogenes, where the original analysis had only identified VKORC1. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 20937004-3 2010 CYP3A4 and CYP3A5 appeared to be the primary enzymes mediating the formation of pharmacologically active ketone metabolite (M1), whereas CYP3A4, CYP3A5, and CYP2C9 appeared to be the enzymes responsible for the formation of the hydroxylated metabolite (M2). Ketones 105-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 20979644-10 2010 Dig 1, non cytotoxic and not inducing caspases by itself, is able to prevent Roundup-induced cell death in a time-dependant manner with an important efficiency of up to 89%, within 48 h. In addition, we evidenced that it prevents Caspases 3/7 activation and CYP3A4 enhancement, and not GST reduction, but in turn it slightly inhibited CYP2C9 when added before Roundup. dig 1 0-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 335-341 20848598-0 2010 Study of atypical kinetic behaviour of cytochrome P450 2C9 isoform with diclofenac at low substrate concentrations by sweeping-MEKC combination. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 20934550-5 2010 Inhibition of CYP2C9 by amiodarone and its major metabolite potentiates the anticoagulant effects of warfarin, increasing the risk of serious bleeding. Amiodarone 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 20934550-5 2010 Inhibition of CYP2C9 by amiodarone and its major metabolite potentiates the anticoagulant effects of warfarin, increasing the risk of serious bleeding. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 20848598-1 2010 In view of the fact that several studies have shown that diclofenac hydroxylation by cytochrome P450 2C9 deviated from Michaelis-Menten kinetics at low substrate concentrations, sweeping combined with MEKC was applied for the kinetic study of this pharmacologically important reaction. Diclofenac 57-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-104 20884456-0 2010 Impact of CYP2C9 genetic polymorphism on warfarin dose requirements in Pakistani population. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 20698928-0 2010 Potential CYP2C9-mediated drug-drug interactions in hospitalized type 2 diabetes mellitus patients treated with the sulphonylureas glibenclamide, glimepiride or glipizide. Glyburide 131-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 20698928-0 2010 Potential CYP2C9-mediated drug-drug interactions in hospitalized type 2 diabetes mellitus patients treated with the sulphonylureas glibenclamide, glimepiride or glipizide. glimepiride 146-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 20698928-0 2010 Potential CYP2C9-mediated drug-drug interactions in hospitalized type 2 diabetes mellitus patients treated with the sulphonylureas glibenclamide, glimepiride or glipizide. Glipizide 161-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glyburide 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glyburide 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glyburide 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. glimepiride 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. glimepiride 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. glimepiride 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glipizide 187-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glipizide 187-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-2 2010 Based on laboratory findings, we determined the clinical significance of potential CYP2C9-mediated drug-drug interactions in hospitalized patients receiving glibenclamide, glimepiride or glipizide, all of which are metabolized by CYP2C9, together with a CYP2C9 inhibitor. Glipizide 187-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 20698928-5 2010 RESULTS: Almost 20% of patients were exposed to a potential drug-drug interaction with a CYP2C9 inhibitor during sulphonylurea treatment. Sulfonylurea Compounds 113-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 20698928-10 2010 CONCLUSIONS: Concomitant use of a CYP2C9 inhibitor results in exaggerated pharmacodynamic effects of sulphonylureas and increases the risk of hypoglycaemia in T2DM patients receiving glibenclamide, glimepiride or glipizide. Sulfonylurea Compounds 101-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20698928-10 2010 CONCLUSIONS: Concomitant use of a CYP2C9 inhibitor results in exaggerated pharmacodynamic effects of sulphonylureas and increases the risk of hypoglycaemia in T2DM patients receiving glibenclamide, glimepiride or glipizide. Glyburide 183-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20698928-10 2010 CONCLUSIONS: Concomitant use of a CYP2C9 inhibitor results in exaggerated pharmacodynamic effects of sulphonylureas and increases the risk of hypoglycaemia in T2DM patients receiving glibenclamide, glimepiride or glipizide. glimepiride 198-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20698928-10 2010 CONCLUSIONS: Concomitant use of a CYP2C9 inhibitor results in exaggerated pharmacodynamic effects of sulphonylureas and increases the risk of hypoglycaemia in T2DM patients receiving glibenclamide, glimepiride or glipizide. Glipizide 213-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20602621-0 2010 Influence of CYP2C9 polymorphism on metabolism of valproate and its hepatotoxin metabolite in Iranian patients. Valproic Acid 50-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 20602621-3 2010 The formation of 4-ene-VPA is catalyzed by cytochrome P450 2C9 (CYP2C9). 2-propyl-4-pentenoic acid 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 20602621-3 2010 The formation of 4-ene-VPA is catalyzed by cytochrome P450 2C9 (CYP2C9). 2-propyl-4-pentenoic acid 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 20884456-2 2010 The objective of our study was to estimate the frequency of genetic and allelic variants of CYP2C9 in Punjabi population of Pakistan and their effects on warfarin dose requirement. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 20884456-10 2010 Individuals with CYP2C9*3/3* need lowest (8.75+-1.76 mg/week) daily warfarin dose. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 20884456-12 2010 Presence of CYP2C9*2 or *3 variants is an independent predictor of low warfarin dose requirement in our patients. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 20642349-1 2010 Voriconazole (VRC), a triazole agent is extensively metabolized by CYP2C19, CYP2C9, and to a lesser extent, by CYP3A4. Voriconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 20602621-4 2010 CYP2C9 allele mutation is closely related to the attenuation of the enzymatic activity and 4-ene-VPA production. 4-ene 91-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20602621-4 2010 CYP2C9 allele mutation is closely related to the attenuation of the enzymatic activity and 4-ene-VPA production. Valproic Acid 97-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20602621-5 2010 In the present work, VPA, 2-ene-VPA, and 4-ene-VPA in serum of patients receiving VPA were determined and the correlation between CYP2C9 polymorphism and 4-ene-VPA concentration was examined. 2-propyl-4-pentenoic acid 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 20602621-15 2010 In conclusion, the role of CYP2C9*2 and CYP2C9*3 in attenuation of 4-ene-VPA formation cannot be confirmed. 4-ene 67-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 20602621-15 2010 In conclusion, the role of CYP2C9*2 and CYP2C9*3 in attenuation of 4-ene-VPA formation cannot be confirmed. 4-ene 67-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 20839301-9 2010 Disappearance of the hydrogen bond between K helix and beta4 loop was observed in CYP2C9*5. Hydrogen 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 20642349-1 2010 Voriconazole (VRC), a triazole agent is extensively metabolized by CYP2C19, CYP2C9, and to a lesser extent, by CYP3A4. Voriconazole 14-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 20877236-4 2010 Eupatilin and jaceosidin potently inhibited CYP1A2-catalyzed phenacetin O-deethylation with 50% inhibitory concentration (IC(50)) values of 9.4 microM and 5.3 microM, respectively, and CYP2C9-catalyzed diclofenac 4-hydroxylation with IC(50) values of 4.1 microM and 10.2 microM, respectively. eupatilin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 20877236-4 2010 Eupatilin and jaceosidin potently inhibited CYP1A2-catalyzed phenacetin O-deethylation with 50% inhibitory concentration (IC(50)) values of 9.4 microM and 5.3 microM, respectively, and CYP2C9-catalyzed diclofenac 4-hydroxylation with IC(50) values of 4.1 microM and 10.2 microM, respectively. jaceosidin 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 20877236-4 2010 Eupatilin and jaceosidin potently inhibited CYP1A2-catalyzed phenacetin O-deethylation with 50% inhibitory concentration (IC(50)) values of 9.4 microM and 5.3 microM, respectively, and CYP2C9-catalyzed diclofenac 4-hydroxylation with IC(50) values of 4.1 microM and 10.2 microM, respectively. Phenacetin 61-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 20877236-6 2010 Kinetic analysis of human liver microsomes showed that eupatilin is a competitive inhibitor of CYP1A2 with a K(i) value of 2.3 microM and a mixed-type inhibitor of CYP2C9 with a K(i) value of 1.6 microM. eupatilin 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 20877236-7 2010 Jaceosidin was shown to be a competitive inhibitor of CYP1A2 with a K(i) value of 3.8 microM and a mixed-type inhibitor of CYP2C9 with K(i) value of 6.4 microM in human liver microsomes. jaceosidin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 20877236-8 2010 These in vitro results suggest that eupatilin and jaceosidin should be further examined for potential pharmacokinetic drug interactions in vivo due to inhibition of CYP1A2 and CYP2C9. eupatilin 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 20877236-8 2010 These in vitro results suggest that eupatilin and jaceosidin should be further examined for potential pharmacokinetic drug interactions in vivo due to inhibition of CYP1A2 and CYP2C9. jaceosidin 50-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 20638257-3 2010 Dihydrotanshinone was a competitive inhibitor of human CYP1A2 (K(i)=0.53 microM) and CYP2C9 (K(i)=1.92 microM), a noncompetitive inhibitor of CYP3A4 (K(i)=2.11 microM) but an uncompetitive CYP2E1 inhibitor. DIHYDROTANSHINONE 0-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 20877452-0 2010 Testing of VKORC1 and CYP2C9 alleles to guide warfarin dosing. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20877452-4 2010 Testing for variants of the vitamin K epoxide reductase complex 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) genes has been proposed for use in guiding the initial dose of warfarin, thus achieving optimal dosing more quickly and with lower risk of bleeding. Warfarin 171-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-98 20877452-4 2010 Testing for variants of the vitamin K epoxide reductase complex 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) genes has been proposed for use in guiding the initial dose of warfarin, thus achieving optimal dosing more quickly and with lower risk of bleeding. Warfarin 171-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 20488169-2 2010 Besides, allele frequencies for clinically relevant genetic markers in warfarin response (i.e., CYP2C9 and VKORC1) have not yet been fully characterized in this population. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 20716240-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT: Variants in the CYP2C9 (i.e. *2 and *3) and VKORC1 (i.e. 1173C/T or -1639G/A) genes have been shown to influence warfarin dose requirements. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 20716240-3 2010 WHAT THIS STUDY ADDS: In African Americans, the VKORC1 rs17886199 variant was statistically significantly associated with log-transformed warfarin maintenance dose, independent of the influence of VKORC1 1173C>T and CYP2C9*2 and *3. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 20668444-4 2010 Whereas glucosamine had no significant effect on CYP activity, noscapine caused marked inhibition of CYP2C9 (4.9-fold increase in urinary losartan/E3174 ratio) and CYP2C19 (3.6-fold increase in the plasma omeprazole/5-hydroxyomeprazole ratio). Noscapine 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 20668444-4 2010 Whereas glucosamine had no significant effect on CYP activity, noscapine caused marked inhibition of CYP2C9 (4.9-fold increase in urinary losartan/E3174 ratio) and CYP2C19 (3.6-fold increase in the plasma omeprazole/5-hydroxyomeprazole ratio). Losartan 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 20668444-4 2010 Whereas glucosamine had no significant effect on CYP activity, noscapine caused marked inhibition of CYP2C9 (4.9-fold increase in urinary losartan/E3174 ratio) and CYP2C19 (3.6-fold increase in the plasma omeprazole/5-hydroxyomeprazole ratio). losartan carboxylic acid 147-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 20668444-5 2010 Noscapine-dependent inhibition of CYP2C9 may explain the interaction with warfarin. Noscapine 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20668444-5 2010 Noscapine-dependent inhibition of CYP2C9 may explain the interaction with warfarin. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20420818-0 2010 Standard warfarin dose in a patient with the CYP2C9*3/*3 genotype leads to hematuria. Warfarin 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 20420818-1 2010 BACKGROUND: Patients with certain CYP2C9 genetic variants have increased sensitivity to warfarin and are at increased risk of over-coagulation with standard warfarin dose. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20420818-1 2010 BACKGROUND: Patients with certain CYP2C9 genetic variants have increased sensitivity to warfarin and are at increased risk of over-coagulation with standard warfarin dose. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 20420818-2 2010 We report over-anticoagulation and hematuria manifest as a slow increase in the international normalized ratio (INR) due to warfarin treatment in a patient with the CYP2C9*3/*3 allele. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 20420818-8 2010 INR response to warfarin in this CYP2C9*3/*3 patient was slow. Warfarin 16-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 20420818-11 2010 In this case, increased risk of bleeding could have been identified by prospective genotyping of CYP2C9 and VKORC1 prior to initiating warfarin therapy. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 20716633-8 2010 Resveratrol intervention was found to inhibit the phenotypic indices of CYP3A4, CYP2D6, and CYP2C9 and to induce the phenotypic index of 1A2. Resveratrol 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 20555338-1 2010 The aim of the study is to improve our understanding of the worldwide allele frequency distribution of four genetic polymorphisms known to influence warfarin dosing (VKORC1 rs9923231, CYP2C9 rs1799853, CYP2C9 rs1057910 and CYP4F2 rs2108622). Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 20555338-1 2010 The aim of the study is to improve our understanding of the worldwide allele frequency distribution of four genetic polymorphisms known to influence warfarin dosing (VKORC1 rs9923231, CYP2C9 rs1799853, CYP2C9 rs1057910 and CYP4F2 rs2108622). Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 202-208 20569971-0 2010 Algorithms using clinical and genetic data (CYP2C9, VKORC1) are relevant to predict warfarin dose in patients with different INR targets. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 27713373-3 2010 The defective alleles of cytochrome P450 (CYP) 2C9 and/or CYP2C19 could affect not only the pharmacokinetics, but also the pharmacodynamics of phenytoin therapy. Phenytoin 143-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-50 20615525-2 2010 Genetic variability in the VKORC1 and CYP2C9 genes is associated with increased warfarin sensitivity. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 20615525-4 2010 VKORC1 and CYP2C9 variability associated with altered warfarin response is less well characterized in African and mixed-raced populations such as Brazilians. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 20615525-5 2010 To determine genetic variability associated with altered warfarin response among Brazilian patients, sixty-two adult patients with extreme resistance or sensitivity to warfarin were genotyped for variants in CYP2C9 and VKORC1. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 20615525-7 2010 Additionally, 52.9% of warfarin sensitive patients had at least one CYP2C9*2 or CYP2C9*3 decreased metabolism allele, 63.6% of Caucasians and 54% of African-descent patients. Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 20615525-7 2010 Additionally, 52.9% of warfarin sensitive patients had at least one CYP2C9*2 or CYP2C9*3 decreased metabolism allele, 63.6% of Caucasians and 54% of African-descent patients. Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 20615525-9 2010 Brazilian patients requiring low doses of warfarin have a high frequency of VKORC1 and CYP2C9 variants associated with warfarin sensitivity. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 20615525-9 2010 Brazilian patients requiring low doses of warfarin have a high frequency of VKORC1 and CYP2C9 variants associated with warfarin sensitivity. Warfarin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 20808793-0 2010 Are cytochrome P450 CYP2C8 and CYP2C9 polymorphisms associated with ibuprofen response in very preterm infants? Ibuprofen 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 20808793-7 2010 Comparing infants with wild type or variant CYP2C8 and CYP2C9 genotypes, response rate to ibuprofen was significantly higher in wild type than in mutated carriers in univariate analysis (73% versus 52%, p = 0.04). Ibuprofen 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 20808793-8 2010 Comparing responders (ductus closure; n = 75) and non-responders (surgical ligation; n = 36), the only two factors significantly associated with the response to ibuprofen using multivariate analysis were higher gestational age and non Caucasian ethnicity but not CYP2C polymorphism. Ibuprofen 161-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 263-268 27713373-5 2010 The defective alleles of CYP2C9 and/or CYP2C19 were also found to have clinically significant effects on the inter-individual variabilities in the population pharmacokinetics of phenobarbital, valproic acid and zonisamide. Phenobarbital 178-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 27713373-5 2010 The defective alleles of CYP2C9 and/or CYP2C19 were also found to have clinically significant effects on the inter-individual variabilities in the population pharmacokinetics of phenobarbital, valproic acid and zonisamide. Valproic Acid 193-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 27713373-5 2010 The defective alleles of CYP2C9 and/or CYP2C19 were also found to have clinically significant effects on the inter-individual variabilities in the population pharmacokinetics of phenobarbital, valproic acid and zonisamide. Zonisamide 211-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 20653669-1 2010 The considerable variability in the warfarin dose-response relationship between individuals, is explained mainly by genetic variation in its major metabolic (CYP2C9) and target (VKORC1) enzymes. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 20653676-5 2010 * This study developed a multiple regression model including CYP2C9, VKORC1 3673G>A, CYP4F2 genotypes and age, weight, combination use of amiodarone which could explain 56.1% of the individual variability in warfarin dose CYP4F2 could explain 4% of the variance in warfarin dose. Warfarin 268-276 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 20653674-0 2010 A haplotype of CYP2C9 associated with warfarin sensitivity in mechanical heart valve replacement patients. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20653674-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * CYP2C9 single nucleotide polymorphisms (SNPs) are important in safe and effective oral anticoagulation with warfarin use. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20653674-2 2010 * Although CYP2C9*2 and *3 are important genetic factors for the warfarin dose, one of the CYP2C9 SNPs, IVS-65G>C, has been suggested to be associated with warfarin sensitivity. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 20653674-2 2010 * Although CYP2C9*2 and *3 are important genetic factors for the warfarin dose, one of the CYP2C9 SNPs, IVS-65G>C, has been suggested to be associated with warfarin sensitivity. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 20653674-2 2010 * Although CYP2C9*2 and *3 are important genetic factors for the warfarin dose, one of the CYP2C9 SNPs, IVS-65G>C, has been suggested to be associated with warfarin sensitivity. Warfarin 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 20405111-0 2010 Fluconazole-induced intoxication with phenytoin in a patient with ultra-high activity of CYP2C9. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 20653674-2 2010 * Although CYP2C9*2 and *3 are important genetic factors for the warfarin dose, one of the CYP2C9 SNPs, IVS-65G>C, has been suggested to be associated with warfarin sensitivity. Warfarin 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 20653674-4 2010 WHAT THIS PAPER ADDS * New information on CYP2C9 SNPs and their occurrences in common haplotype structures in healthy unrelated Koreans and in individuals who require low warfarin dose after mechanical heart valve replacements (MHVRs) were studied. Warfarin 171-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 20653674-8 2010 Additional direct DNA sequencing of the CYP2C9 gene was conducted in 36 of the 267 MHVR patients who required low maintenance warfarin doses without carrying CYP2C9*3 and VKORC1 1173T mutations. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 20653674-9 2010 The effects of CYP2C9 genetics on warfarin maintenance dose were assessed in 267 MHVR patients. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20653674-11 2010 One of the CYP2C9 haplotypes exhibited an association with warfarin low dose requirement. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 20653676-1 2010 WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Genetic polymorphisms of VKORC1 and CYP2C9 are known to influence warfarin dosage. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 20653676-5 2010 * This study developed a multiple regression model including CYP2C9, VKORC1 3673G>A, CYP4F2 genotypes and age, weight, combination use of amiodarone which could explain 56.1% of the individual variability in warfarin dose CYP4F2 could explain 4% of the variance in warfarin dose. Amiodarone 141-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 20653676-5 2010 * This study developed a multiple regression model including CYP2C9, VKORC1 3673G>A, CYP4F2 genotypes and age, weight, combination use of amiodarone which could explain 56.1% of the individual variability in warfarin dose CYP4F2 could explain 4% of the variance in warfarin dose. Warfarin 211-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 20501911-3 2010 The kinetics of metabolite formation from phenacetin, tolbutamide, and alprazolam and midazolam, selected as substrates probes for CYP1A2, CYP2C9, and CYP3A4, respectively, were characterized in this in vitro system. Phenacetin 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 20501911-3 2010 The kinetics of metabolite formation from phenacetin, tolbutamide, and alprazolam and midazolam, selected as substrates probes for CYP1A2, CYP2C9, and CYP3A4, respectively, were characterized in this in vitro system. Alprazolam 71-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 20501911-3 2010 The kinetics of metabolite formation from phenacetin, tolbutamide, and alprazolam and midazolam, selected as substrates probes for CYP1A2, CYP2C9, and CYP3A4, respectively, were characterized in this in vitro system. Midazolam 86-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 20405111-0 2010 Fluconazole-induced intoxication with phenytoin in a patient with ultra-high activity of CYP2C9. Phenytoin 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 20405111-1 2010 PURPOSE: The cytochrome P450 enzyme CYP2C9 metabolizes several important drugs, such as warfarin and oral antidiabetic drugs. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 20405111-5 2010 When fluconazole, a potent inhibitor of CYP2C9, was added to the treatment regimen, the patient developed ataxia, tremor, fatigue, slurred speech and somnolence, indicating phenytoin intoxication. Fluconazole 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 20405111-5 2010 When fluconazole, a potent inhibitor of CYP2C9, was added to the treatment regimen, the patient developed ataxia, tremor, fatigue, slurred speech and somnolence, indicating phenytoin intoxication. Phenytoin 173-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 20405111-6 2010 On suspicion of ultra-high activity of CYP2C9, a phenotyping test for CYP2C9 with losartan was performed. Losartan 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 20405111-8 2010 CONCLUSIONS: Our finding of an ultrarapid metabolism of losartan and phenytoin may apply to other CYP2C9 substrates, where inhibition of CYP2C9 may cause severe adverse drug reactions. Losartan 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 20405111-8 2010 CONCLUSIONS: Our finding of an ultrarapid metabolism of losartan and phenytoin may apply to other CYP2C9 substrates, where inhibition of CYP2C9 may cause severe adverse drug reactions. Losartan 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20405111-8 2010 CONCLUSIONS: Our finding of an ultrarapid metabolism of losartan and phenytoin may apply to other CYP2C9 substrates, where inhibition of CYP2C9 may cause severe adverse drug reactions. Phenytoin 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 20405111-8 2010 CONCLUSIONS: Our finding of an ultrarapid metabolism of losartan and phenytoin may apply to other CYP2C9 substrates, where inhibition of CYP2C9 may cause severe adverse drug reactions. Phenytoin 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20509749-3 2010 In liver microsomes from PB-treated pigs, these transcriptional activations were accompanied by an increase of various marker activities of human CYP2B6, CYP3As, CYP2C9, CYP2C19. Phenobarbital 25-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 20135639-7 2010 In incubations with human liver microsomes and selective inhibitors, CYP2E1 was found to be principally responsible for producing the dominant, hydroxylation product, whereas CYP2C9 was the principal source of the epoxides and CYP1A2 was responsible for the dealkylation product. Epoxy Compounds 214-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 20509749-4 2010 Among the extrahepatic tissues, a significant induction by PB was observed only in kidney for the marker activities of CYP2C9. Phenobarbital 59-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 20400659-6 2010 The current study compares the application of such d-optimal designs with that of a conventional approach in assessing the competitive inhibitory potency of fluconazole and sertraline toward CYP2C9 and 2D6, respectively. Fluconazole 157-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 20677151-0 2010 [Association of polymorphisms of cytochrome P450 2C9 exon 4 and -65G>C with warfarin sensitivity]. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-52 20677151-1 2010 OBJECTIVE: To investigate the association of the polymorphisms of cytochrome P450 2C9 (CYP2C9) exon 4 608T/G, 561A/C, 537A/C and 527A/C, and -65G/C with warfarin sensitivity. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-85 20677151-1 2010 OBJECTIVE: To investigate the association of the polymorphisms of cytochrome P450 2C9 (CYP2C9) exon 4 608T/G, 561A/C, 537A/C and 527A/C, and -65G/C with warfarin sensitivity. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 20677151-10 2010 Warfarin daily dosage in CYP2C9 exon 4 -65C carriers was 3.106+/-0.619 mg/d, while it was (2.555+/-0.708) mg/d in individuals with wild-type -65G (P=0.020). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 20677151-11 2010 Receiver operating characteristic (ROC) analysis showed that warfarin daily dosage of more than 2.5 mg/d can be used to predict the CYP2C9 exon 4 -65GC genotype (AUC: 0.770, P=0.005, 95%CI:0.626-0.915). Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 20677151-15 2010 The warfarin maintenance dosage in CYP2C9 exon 4 -65CG carriers was significantly higher than those with wild-type -65GG. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 20733952-1 2010 Genetic testing for common variants in the CYP2C9 and VKORC1 genes may provide useful clinical information to guide dosing patients receiving oral warfarin. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20733952-2 2010 Specifically, the CYP2C9*2, CYP2C9*3 and either the VKORC1-1639 G>A or VKORC1 1173C>T polymorphisms can be used to help predict an approximate warfarin maintenance dose needed for a particular patient. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 20733952-2 2010 Specifically, the CYP2C9*2, CYP2C9*3 and either the VKORC1-1639 G>A or VKORC1 1173C>T polymorphisms can be used to help predict an approximate warfarin maintenance dose needed for a particular patient. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 20210733-0 2010 Distribution of CYP2C9 and VKORC1 risk alleles for warfarin sensitivity and resistance in the Israeli population. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 20210733-1 2010 PURPOSE: This study was designed to delineate the relative frequency of CYP2C9 and VKORC1 polymorphisms known to affect warfarin response in the highly heterogeneous Israeli population. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 20210733-4 2010 RESULTS: Arab Moslems had a higher frequency of warfarin "sensitive" CYP2C9*2, CYP2C9*3 and VKROC1 -1639G>A alleles (0.21, 0.07 and 0.58, respectively) than both Jews (0.13, 0.11 and 0.57, respectively) and Druze (0.12, 0.06 and 0.53, respectively). Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20210733-8 2010 The results presented demonstrate that allelic variants in CYP2C9 and VKORC1 are very common in Israel with approximately 95% of Jews, approximately 84% of Druze and approximately 91% of Arab Moslems manifesting at least one known warfarin "sensitive" or "resistant" allele. Warfarin 231-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 20226775-0 2010 Comparison of assay systems for warfarin-related CYP2C9 and VKORC1 genotyping. Warfarin 32-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 20400659-6 2010 The current study compares the application of such d-optimal designs with that of a conventional approach in assessing the competitive inhibitory potency of fluconazole and sertraline toward CYP2C9 and 2D6, respectively. Sertraline 173-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 20390258-0 2010 Influence of CYP2C9 and CYP2C19 genetic polymorphisms on phenytoin-induced neurological toxicity in Indian epileptic patients. Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 20390258-9 2010 CONCLUSION: Our results show that CYP2C9 genetic polymorphisms (particularly the *3 allele) were associated with high risk of epileptic patients developing PHT-induced neurological toxicity. Phenytoin 156-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. Tolbutamide 57-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 20162308-11 2010 Down regulation of CYP2C9 and CYP2C19 synthesis by 5FU therapies may explain the adverse effect of 5FU on the clinical disposition of warfarin and phenytoin. Fluorouracil 51-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 20162308-11 2010 Down regulation of CYP2C9 and CYP2C19 synthesis by 5FU therapies may explain the adverse effect of 5FU on the clinical disposition of warfarin and phenytoin. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 20162308-11 2010 Down regulation of CYP2C9 and CYP2C19 synthesis by 5FU therapies may explain the adverse effect of 5FU on the clinical disposition of warfarin and phenytoin. Phenytoin 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 20133511-2 2010 Treprostinil is metabolized primarily by cytochrome P450 (CYP) 2C8 with minor contribution from CYP2C9. treprostinil 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 20133511-3 2010 It is expected that oral treprostinil will be administered with bosentan, approved for the treatment of PAH and known to induce CYP2C9 and 3A4. treprostinil 25-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 20133511-3 2010 It is expected that oral treprostinil will be administered with bosentan, approved for the treatment of PAH and known to induce CYP2C9 and 3A4. Bosentan 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 20865058-2 2010 The mechanism could involve inhibition of the hepatic CYP2C9-mediated metabolic clearance of warfarin by components in cranberry. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 20865058-4 2010 METHODS: The inhibitory effects of five types of cranberry juices were compared with those of water on CYP2C9 activity (S-warfarin 7-hydroxylation) in human liver microsomes (HLM). Water 94-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 20865058-4 2010 METHODS: The inhibitory effects of five types of cranberry juices were compared with those of water on CYP2C9 activity (S-warfarin 7-hydroxylation) in human liver microsomes (HLM). Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. Tolbutamide 44-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. Tolbutamide 44-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. Tolbutamide 57-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. 4'-hydroxytolbutamide 64-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. 4'-hydroxytolbutamide 64-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. 4-oh-tb 87-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23964176-2 2010 METHOD: CYP2C9-mediated 4"-hydroxylation of tolbutamide (TB) to 4"-hydroxytolbutamide (4-OH-TB) was utilized to assess the metabolic activity of CYP2C9. 4-oh-tb 87-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 23964176-8 2010 CONCLUSION: The results of this study have shown that alcoholic extracts of curcuma and aniseed were capable of inhibiting and activating; respectively, the CYP2C9-mediated 4-OH-TB formation in human liver microsomes, suggesting that these herbs have the potential to interact with CYP2C9 drug substrates. 4-oh-tb 173-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 23964176-8 2010 CONCLUSION: The results of this study have shown that alcoholic extracts of curcuma and aniseed were capable of inhibiting and activating; respectively, the CYP2C9-mediated 4-OH-TB formation in human liver microsomes, suggesting that these herbs have the potential to interact with CYP2C9 drug substrates. 4-oh-tb 173-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 282-288 20393654-1 2010 HyBeacon probes have been used to characterise SNPs in the CYP2C9 and VKORC1 genes associated with variations in the efficiency of warfarin metabolism. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 20381283-2 2010 BACKGROUND: Genotypic variations in CYP2C9 and VKORC1 have been shown to predict warfarin dosing, but no large-scale studies have prospectively evaluated the clinical effectiveness of genotyping in naturalistic settings across the U.S. METHODS: This national, prospective, comparative effectiveness study compared the 6-month incidence of hospitalization in patients receiving warfarin genotyping (n = 896) versus a matched historical control group (n = 2,688). Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 20436375-2 2010 CYP2C enzymes catalyze the metabolism of arachidonic acid to EETs. Arachidonic Acid 41-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-5 20442691-0 2010 Extending and evaluating a warfarin dosing algorithm that includes CYP4F2 and pooled rare variants of CYP2C9. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 20442691-10 2010 CONCLUSION: CYP4F2 and pooled rare variants of CYP2C9 significantly improve the ability to estimate warfarin dose. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 20585445-0 2010 A novel, single algorithm approach to predict acenocoumarol dose based on CYP2C9 and VKORC1 allele variants. Acenocoumarol 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 20585445-1 2010 The identification of CYP2C9 and VKORC1 genes has strongly stimulated the research on pharmacogenetics of coumarins in the last decade. Coumarins 106-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20376629-2 2010 Polymorphisms in the vitamin K epoxide reductase complex subunit 1 (VKORC1) and the liver enzyme cytochrome P450 2C9 (CYP2C9) effectively account for the variability in warfarin and acenocoumarol response but are less well-defined pharmacogenetic predictors in phenprocoumon therapy. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-116 20410877-0 2010 A pharmacometric model describing the relationship between warfarin dose and INR response with respect to variations in CYP2C9, VKORC1, and age. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 20445534-0 2010 CYP2C9*3 Loss-of-Function Allele Is Associated With Acute Upper Gastrointestinal Bleeding Related to the Use of NSAIDs Other Than Aspirin. Aspirin 130-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20445534-1 2010 Nonsteroidal anti-inflammatory drugs (NSAIDs), other than aspirin, are to some extent metabolized by cytochrome P450 2C9 (CYP2C9). Aspirin 58-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-120 20445534-1 2010 Nonsteroidal anti-inflammatory drugs (NSAIDs), other than aspirin, are to some extent metabolized by cytochrome P450 2C9 (CYP2C9). Aspirin 58-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 20445534-5 2010 In the aspirin group, 12 patients (9.2%) had the CYP2C9 359Leu allele as compared with 19 (33.3%) in the non-ASP group (odds ratio (OR) = 5.0; 95% confidence interval 2.2-11.1, P < 0.0001). Aspirin 7-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 20445534-6 2010 In a multivariate analysis, CYP2C9 359Leu remained associated with the non-ASP group (OR = 7.2 (2.6-20.3), P = 0.0002) even though 40% of these patients were under treatment with antiulcer drugs at the time of admission. Aspartic Acid 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 20215413-4 2010 CYP2C9-mediated metabolism of S-naproxen and S-flurbiprofen was inhibited up to 80% by coincubation with CYP3A4, although K(m) values were unchanged. Naproxen 30-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20215413-4 2010 CYP2C9-mediated metabolism of S-naproxen and S-flurbiprofen was inhibited up to 80% by coincubation with CYP3A4, although K(m) values were unchanged. Flurbiprofen 45-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20637959-15 2010 Multiple linear regression analysis found that sex (P = 0.045), height (P = 0.024), age (P = 0.081), and VKORC1 (P = 0.004) and CYP2C9 (P = 0.011) polymorphism had significant influence on the maintenance dose of warfarin. Warfarin 213-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 20637959-18 2010 CONCLUSION: Among the demographic and genetic factors evaluated in these Iranian patients, sex, height, age, CYP2C9, and VKORC1 had significant effects on warfarin maintenance dose requirements. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 20376629-2 2010 Polymorphisms in the vitamin K epoxide reductase complex subunit 1 (VKORC1) and the liver enzyme cytochrome P450 2C9 (CYP2C9) effectively account for the variability in warfarin and acenocoumarol response but are less well-defined pharmacogenetic predictors in phenprocoumon therapy. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 20376629-2 2010 Polymorphisms in the vitamin K epoxide reductase complex subunit 1 (VKORC1) and the liver enzyme cytochrome P450 2C9 (CYP2C9) effectively account for the variability in warfarin and acenocoumarol response but are less well-defined pharmacogenetic predictors in phenprocoumon therapy. Acenocoumarol 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-116 20376629-2 2010 Polymorphisms in the vitamin K epoxide reductase complex subunit 1 (VKORC1) and the liver enzyme cytochrome P450 2C9 (CYP2C9) effectively account for the variability in warfarin and acenocoumarol response but are less well-defined pharmacogenetic predictors in phenprocoumon therapy. Acenocoumarol 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 20831536-0 2010 Influence of CYP2C9 and CYP2C19 genetic polymorphisms on pharmacokinetics and pharmacodynamics of gliclazide in healthy Chinese Han volunteers. Gliclazide 98-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 20831536-1 2010 BACKGROUND AND OBJECTIVE: CYP2C9 is the major contributor to gliclazide metabolic clearance in vitro, while the pharmacokinetics of gliclazide modified release are affected mainly by CYP2C19 genetic polymorphisms in vivo. Gliclazide 61-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 20831536-2 2010 This study aims to investigate the influence of CYP2C9 and CYP2C19 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of gliclazide in healthy Chinese Han volunteers. Gliclazide 137-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 20429590-0 2010 Hydroxywarfarin metabolites potently inhibit CYP2C9 metabolism of S-warfarin. hydroxywarfarin 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 20386359-2 2010 The relationship between warfarin sensitivity and CYP2C9 and VKORC1 variants is strong, and is the basis for several proposed dosing algorithms. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 20386359-11 2010 In addition, the results of this study confirm those of others demonstrating clear relationship of genotype for CYP2C9 and VKORC1 with warfarin dose requirements; as the number of variants in these genes increases, the dose requirement decreases. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 20421126-0 2010 A regression model to predict warfarin dose from clinical variables and polymorphisms in CYP2C9, CYP4F2, and VKORC1: Derivation in a sample with predominantly a history of venous thromboembolism. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 20429590-0 2010 Hydroxywarfarin metabolites potently inhibit CYP2C9 metabolism of S-warfarin. Warfarin 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 20429590-5 2010 We hypothesized that hydroxywarfarins inhibit the hydroxylation of warfarin by CYP2C9, thereby limiting enzymatic capacity toward S-warfarin. hydroxywarfarins 21-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 20429590-5 2010 We hypothesized that hydroxywarfarins inhibit the hydroxylation of warfarin by CYP2C9, thereby limiting enzymatic capacity toward S-warfarin. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 20429590-5 2010 We hypothesized that hydroxywarfarins inhibit the hydroxylation of warfarin by CYP2C9, thereby limiting enzymatic capacity toward S-warfarin. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 20429590-6 2010 To test this hypothesis, we investigated the ability of all five racemic hydroxywarfarins to block CYP2C9 activity toward S-warfarin using recombinant enzyme and human liver microsomes. hydroxywarfarins 73-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 20429590-6 2010 To test this hypothesis, we investigated the ability of all five racemic hydroxywarfarins to block CYP2C9 activity toward S-warfarin using recombinant enzyme and human liver microsomes. Sulfur 122-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 20429590-6 2010 To test this hypothesis, we investigated the ability of all five racemic hydroxywarfarins to block CYP2C9 activity toward S-warfarin using recombinant enzyme and human liver microsomes. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 20429590-7 2010 We initially screened for the inhibition of CYP2C9 by hydroxywarfarins using a P450-Glo assay to determine IC(50) values for each hydroxywarfarin. hydroxywarfarins 54-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 20429590-7 2010 We initially screened for the inhibition of CYP2C9 by hydroxywarfarins using a P450-Glo assay to determine IC(50) values for each hydroxywarfarin. hydroxywarfarin 54-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 20429590-8 2010 Compared to the substrate, CYP2C9 bound its hydroxywarfarin products with less affinity but retained high affinity for 10- and 4"-hydroxywarfarins, products from CYP3A4 reactions. hydroxywarfarin 44-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 20429590-8 2010 Compared to the substrate, CYP2C9 bound its hydroxywarfarin products with less affinity but retained high affinity for 10- and 4"-hydroxywarfarins, products from CYP3A4 reactions. 10- and 4"-hydroxywarfarins 119-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 20429590-9 2010 S-Warfarin steady-state inhibition studies with recombinant CYP2C9 and pooled human liver microsomes confirmed that hydroxywarfarin products from CYP reactions possess the capacity to competitively inhibit CYP2C9 with biologically relevant inhibition constants. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 20429590-9 2010 S-Warfarin steady-state inhibition studies with recombinant CYP2C9 and pooled human liver microsomes confirmed that hydroxywarfarin products from CYP reactions possess the capacity to competitively inhibit CYP2C9 with biologically relevant inhibition constants. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 20429590-9 2010 S-Warfarin steady-state inhibition studies with recombinant CYP2C9 and pooled human liver microsomes confirmed that hydroxywarfarin products from CYP reactions possess the capacity to competitively inhibit CYP2C9 with biologically relevant inhibition constants. hydroxywarfarin 116-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 20429590-9 2010 S-Warfarin steady-state inhibition studies with recombinant CYP2C9 and pooled human liver microsomes confirmed that hydroxywarfarin products from CYP reactions possess the capacity to competitively inhibit CYP2C9 with biologically relevant inhibition constants. hydroxywarfarin 116-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 20429590-10 2010 Inhibition of CYP2C9 by 7-hydroxywarfarin may be significant given its abundance in human plasma, despite its weak affinity for the enzyme. 7-hydroxywarfarin 24-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 20429590-11 2010 10-Hydroxywarfarin, which has been reported as the second most abundant plasma metabolite, was the most potent inhibitor of CYP2C9, displaying approximately 3-fold higher affinity than S-warfarin. 10-hydroxywarfarin 0-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 20429590-11 2010 10-Hydroxywarfarin, which has been reported as the second most abundant plasma metabolite, was the most potent inhibitor of CYP2C9, displaying approximately 3-fold higher affinity than S-warfarin. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 20429590-12 2010 These results indicate that hydroxywarfarin metabolites produced by CYP2C9 and other CYPs may limit metabolic capacity toward S-warfarin through competitive inhibition. hydroxywarfarin 28-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 20429590-12 2010 These results indicate that hydroxywarfarin metabolites produced by CYP2C9 and other CYPs may limit metabolic capacity toward S-warfarin through competitive inhibition. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 20375999-1 2010 Well-characterized genes that affect warfarin metabolism (cytochrome P450 (CYP) 2C9) and sensitivity (vitamin K epoxide reductase complex 1 (VKORC1)) explain one-third of the variability in therapeutic dose before the international normalized ratio (INR) is measured. Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-83 20437462-5 2010 The present study systematically investigated the contributions of various human CYP isoforms (CYP3A4, CYP3A5, CYP2C8, CYP2C9 and CYP2C19) to in vitro metabolism of GLB. Glyburide 165-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 20203262-1 2010 Warfarin-dosing algorithms incorporating CYP2C9 and VKORC1 -1639G>A improve dose prediction compared with algorithms based solely on clinical and demographic factors. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 20089352-0 2010 The influence of cytochrome oxidase CYP2A6, CYP2B6, and CYP2C9 polymorphisms on the plasma concentrations of valproic acid in epileptic patients. Valproic Acid 109-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 20375999-3 2010 After adjusting for INR, CYP2C9 and VKORC1 genotypes remained significant predictors (P < 0.001) of warfarin dose. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 20609060-1 2010 OBJECTIVES: Nateglinide is metabolized by CYP2C9 and CYP3A4, therefore drug-drug interactions through cytochrome P450 (CYP) inhibition may occur. Nateglinide 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 20354686-0 2010 Increased frequency of CYP2C9 variant alleles and homozygous VKORC1*2B carriers in warfarin-treated patients with excessive INR response. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 20354686-1 2010 BACKGROUND: Several studies have linked mutations in the genes encoding cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) to a reduced warfarin dose requirement and an increased risk of bleeding with warfarin treatment, but the implementation of genotyping as routine practice is still controversial. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-91 20354686-1 2010 BACKGROUND: Several studies have linked mutations in the genes encoding cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) to a reduced warfarin dose requirement and an increased risk of bleeding with warfarin treatment, but the implementation of genotyping as routine practice is still controversial. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 20354686-1 2010 BACKGROUND: Several studies have linked mutations in the genes encoding cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) to a reduced warfarin dose requirement and an increased risk of bleeding with warfarin treatment, but the implementation of genotyping as routine practice is still controversial. Warfarin 230-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-91 20354686-1 2010 BACKGROUND: Several studies have linked mutations in the genes encoding cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) to a reduced warfarin dose requirement and an increased risk of bleeding with warfarin treatment, but the implementation of genotyping as routine practice is still controversial. Warfarin 230-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 20354686-2 2010 The objective of this study was to investigate whether the frequencies of CYP2C9 variant alleles (*2 and *3), and VKORC1 haplotypes (*2A/B) were increased in a population of warfarin-treated patients with an excessive INR response. Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 20354686-10 2010 CONCLUSION: The presence of CYP2C9 variant alleles and the homozygous VKORC1*2B genotype was associated with elevated INR values in warfarin-treated patients. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 20609060-4 2010 KEY FINDINGS: The results showed that nateglinide inhibited CYP2C9 and CYP2C19 with an IC50(app) (apparent value of the 50% inhibitory concentration) of 125 micromol/l and 946 micromol/l, respectively, while M1 did not inhibit any of the CYP isoforms. Nateglinide 38-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 20609060-5 2010 The inhibition constant (K(i)) value of the inhibitory effect of nateglinide on CYP2C9 and the 1 + I(in,max,u)/K(i) value were estimated (where I(in,max,u)= the maximum unbound concentration of nateglinide). Nateglinide 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 18998206-5 2010 Polymorphic sites in three genes, cytochrome P450 (CYP) 2C9, vitamin K 2,3 epoxide reductase complex 1 (VKORC1), and CYP4F2, have been shown to affect stable warfarin dose. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-59 20609060-7 2010 The influence of pre-incubation on the inhibition by nateglinide of CYP3A4, CYP2C9 and CYP2C19 was examined. Nateglinide 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 20609060-3 2010 METHODS: We used typical substrates (7-ethoxyresorufin for CYP1A1/2, tolbutamide for CYP2C9, S-mephenytoin for CYP2C19, bufuralol for CYP2D6, chlorzoxazone for CYP2E1 and midazolam for CYP3A4) in the evaluation of the inhibitory effects, and examined the possibility of mechanism-based inhibition (MBI) by evaluating the influence of pre-incubation in the inhibition. Tolbutamide 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 20149073-12 2010 CONCLUSION: Two clinical data, age and BSA, and three SNPs in the VKORC1, CYP2C9 and CYP4F2 genes strongly predict outlier patients treated with acenocoumarol. Acenocoumarol 145-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 20086032-3 2010 In the present study, we used primary cultures of human hepatocytes to investigate the role of HNF4alpha in the pregnane X receptor (PXR)/rifampicin-mediated up-regulation of CYP2C8, CYP2C9, and CYP2C19 gene expression. Rifampin 138-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 29776219-1 2010 AIMS: Patient demographics and variant alleles in the CYP2C9 and VKORC1 genes account for 50% of the population variability in warfarin maintenance doses. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 20072124-2 2010 In a cohort of 226 African-American patients, weekly warfarin dose requirements were lower in those with the CYP2C9*8 allele (34 (30-47) mg; P = 0.023) and the CYP2C9 *2, *3, *5, *6, or *11 allele (33(28-40 mg); P < 0.001) as compared with those with the CYP2C9*1/*1 genotype (43 (35-56) mg). Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 20072124-2 2010 In a cohort of 226 African-American patients, weekly warfarin dose requirements were lower in those with the CYP2C9*8 allele (34 (30-47) mg; P = 0.023) and the CYP2C9 *2, *3, *5, *6, or *11 allele (33(28-40 mg); P < 0.001) as compared with those with the CYP2C9*1/*1 genotype (43 (35-56) mg). Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 20072124-2 2010 In a cohort of 226 African-American patients, weekly warfarin dose requirements were lower in those with the CYP2C9*8 allele (34 (30-47) mg; P = 0.023) and the CYP2C9 *2, *3, *5, *6, or *11 allele (33(28-40 mg); P < 0.001) as compared with those with the CYP2C9*1/*1 genotype (43 (35-56) mg). Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 20072124-3 2010 The combination of CYP2C9 alleles, VKORC1 -1639G>A genotype, and clinical variables explained 36% of the interpatient variability in warfarin dose requirements. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 20072124-6 2010 The inclusion of additional CYP2C9 variants may improve the predictive ability of warfarin dosing algorithms for African Americans. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 20086032-7 2010 Mutational studies showed that the HNF4alpha sites are needed for up-regulation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 20086032-8 2010 Furthermore, silencing of HNF4alpha abolished transactivation of the CYP2C8 and CYP2C9 promoters by rifampicin. Rifampin 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 20586862-5 2010 Carrying the CYP2C9 variants is reported a significantly increased risk of non-aspirin NSAID-related GI bleeding. Aspirin 79-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19960413-0 2010 Inhibitory effects of deoxypodophyllotoxin from Anthriscus sylvestris on human CYP2C9 and CYP3A4. deoxypodophyllotoxin 22-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19960413-3 2010 When a mixture of specific CYP substrates was incubated with DPT in HLM, CYP2C9-catalyzed diclofenac 4-hydroxylation and CYP3A4-catalyzed midazolam 1-hydroxylation were strongly inhibited by DPT, with IC (50) values of 6.3 and 9.2 microM, respectively. deoxypodophyllotoxin 61-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19960413-3 2010 When a mixture of specific CYP substrates was incubated with DPT in HLM, CYP2C9-catalyzed diclofenac 4-hydroxylation and CYP3A4-catalyzed midazolam 1-hydroxylation were strongly inhibited by DPT, with IC (50) values of 6.3 and 9.2 microM, respectively. Diclofenac 90-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19960413-3 2010 When a mixture of specific CYP substrates was incubated with DPT in HLM, CYP2C9-catalyzed diclofenac 4-hydroxylation and CYP3A4-catalyzed midazolam 1-hydroxylation were strongly inhibited by DPT, with IC (50) values of 6.3 and 9.2 microM, respectively. deoxypodophyllotoxin 191-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19960413-5 2010 From these results, DPT was characterized to be a competitive inhibitor of CYP2C9 and CYP3A4, with K(i) values of 3.5 and 10.8 microM in HLM and 24.9 and 3.5 microM in baculovirus-insect cell-expressed human CYPs, respectively. deoxypodophyllotoxin 20-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 20200517-1 2010 Warfarin demonstrates a wide interindividual variability in response that is mediated partly by variants in cytochrome P450 2C9 (CYP2C9) and vitamin K 2,3-epoxide reductase complex subunit 1 (VKORC1). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-127 20200517-1 2010 Warfarin demonstrates a wide interindividual variability in response that is mediated partly by variants in cytochrome P450 2C9 (CYP2C9) and vitamin K 2,3-epoxide reductase complex subunit 1 (VKORC1). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 19562511-1 2010 Acenocoumarol is mainly catabolized by CYP2C9 isoform of cytochrome P450 (CYP) liver complex and exerts its anticoagulant effect through the inhibition of Vitamin K Epoxide Reductase (VKOR). Acenocoumarol 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 19562511-2 2010 The most important genetic polymorphisms which lead to an impaired enzymatic activity and therefore predispose to acenocoumarol sensitivity, are considered to be CYP2C9*2 (Arg144Cys), CYP2C9*3 (Ile359Leu) and VKORC1-1639G>A, respectively. Acenocoumarol 114-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19562511-2 2010 The most important genetic polymorphisms which lead to an impaired enzymatic activity and therefore predispose to acenocoumarol sensitivity, are considered to be CYP2C9*2 (Arg144Cys), CYP2C9*3 (Ile359Leu) and VKORC1-1639G>A, respectively. Acenocoumarol 114-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 19562511-2 2010 The most important genetic polymorphisms which lead to an impaired enzymatic activity and therefore predispose to acenocoumarol sensitivity, are considered to be CYP2C9*2 (Arg144Cys), CYP2C9*3 (Ile359Leu) and VKORC1-1639G>A, respectively. ile359leu 194-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19562511-2 2010 The most important genetic polymorphisms which lead to an impaired enzymatic activity and therefore predispose to acenocoumarol sensitivity, are considered to be CYP2C9*2 (Arg144Cys), CYP2C9*3 (Ile359Leu) and VKORC1-1639G>A, respectively. ile359leu 194-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 19347265-1 2010 Seven fungal cultures were studied for the metabolism of diclofenac in order to elucidate the nature of enzymes involved in biotransformation, as diclofenac is a specific substrate to cytochrome P450 (CYP) 2C9 isozyme in mammals. Diclofenac 57-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-209 20350128-3 2010 Several retrospective and a few small prospective clinical studies have shown that polymorphisms in CYP2C9 and VKORC1 genes together account for 35-50% of the variability in warfarin initiation and maintenance dose requirements. Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 19942260-2 2010 It has been suggested that anticoagulation effect of warfarin is significantly associated with the polymorphism of certain genes, including Cytochrome P450 complex subunit 2C9 (CYP2C9), Vitamin K Epoxide Reductase Complex Subunit 1 (VKORC1), Gamma-Glutamyl Carboxylase (GGCX) and Apolipoprotein E (APOE) etc. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-175 19942260-2 2010 It has been suggested that anticoagulation effect of warfarin is significantly associated with the polymorphism of certain genes, including Cytochrome P450 complex subunit 2C9 (CYP2C9), Vitamin K Epoxide Reductase Complex Subunit 1 (VKORC1), Gamma-Glutamyl Carboxylase (GGCX) and Apolipoprotein E (APOE) etc. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 19347265-3 2010 The study included clopidogrel, a selective inhibitor of CYP2C9 isozyme, to inhibit the metabolism of diclofenac. Clopidogrel 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 19347265-3 2010 The study included clopidogrel, a selective inhibitor of CYP2C9 isozyme, to inhibit the metabolism of diclofenac. Diclofenac 102-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 19995889-1 2010 CYP2C9 is an important drug-metabolizing enzyme that metabolizes, e.g., warfarin, antidiabetics, and antiphlogistics. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20178046-5 2010 Fluvastatin, which is metabolized by CYP2C9, is less prone to pharmacokinetic interactions, while pravastatin, rosuvastatin and pitavastatin are not susceptible to any CYP inhibition. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 20339978-4 2010 The mean warfarin daily dose requirement was higher in CYP2C9 homozygous wild-type patients, compared to those with the variant *3 allele (P < 0.05), similar to those with the variant *2 allele (P > 0.05) and highest in patients with the VKORC1 -1639 GG genotype compared to those with the GA genotype and the AA genotype (P < 0.01). Warfarin 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 20020283-7 2010 A multiple regression model including age, sex, last INR, VKORC1, and CYP2C9 genotypes explained ~50% of the variability in AC/PC dose requirements. Acenocoumarol 124-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 20020283-7 2010 A multiple regression model including age, sex, last INR, VKORC1, and CYP2C9 genotypes explained ~50% of the variability in AC/PC dose requirements. Phenprocoumon 127-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 20222813-4 2010 In this article, we review a study examining the association of genetic variation in the cytochrome P450 2C9 enzyme with glycemic response to sulfonylureas in a large cohort of patients with Type 2 diabetes from the Genetics of Diabetes Audit and Research Tayside Study (Go-DARTS). Sulfonylurea Compounds 142-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-108 20339978-7 2010 VKORC1 genotype and CYP2C9 polymorphism affect daily dose requirements and time to therapeutic INR in Turkish patients receiving warfarin for anticoagulation. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 20402581-2 2010 Variations in two genes, VKORC1 and CYP2C9, have been associated with variation in warfarin metabolism among individuals. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 20093140-4 2010 CYP epoxygenases such as CYP1A2, CYP2C, and CYP2J2 metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which generally possess vasodilating, anti-inflammatory, anti-apoptotic, anti-thrombotic, natriuretic, and cardioprotective effects. Arachidonic Acid 62-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-38 19934397-0 2010 Two flavonolignans from milk thistle (Silybum marianum) inhibit CYP2C9-mediated warfarin metabolism at clinically achievable concentrations. Flavonolignans 4-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 19934397-0 2010 Two flavonolignans from milk thistle (Silybum marianum) inhibit CYP2C9-mediated warfarin metabolism at clinically achievable concentrations. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 20402581-5 2010 The prevalence of CYP2C9 and VKORC1 polymorphisms vary among different ethnic groups, and can account for over 30% of variance in warfarin dose. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 20233183-10 2010 CONCLUSIONS: TDI of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 20169036-4 2010 Etravirine is a substrate of CYP3A4, CYP2C9, CYP2C19, and uridine diphosphate glucuronyltransferase, and induces CYP3A4, weakly inhibits CYP2C9 and moderately inhibits CYP2C19. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 20169036-4 2010 Etravirine is a substrate of CYP3A4, CYP2C9, CYP2C19, and uridine diphosphate glucuronyltransferase, and induces CYP3A4, weakly inhibits CYP2C9 and moderately inhibits CYP2C19. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20233183-0 2010 Time-dependent inhibition (TDI) of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Noscapine 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 20093140-4 2010 CYP epoxygenases such as CYP1A2, CYP2C, and CYP2J2 metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which generally possess vasodilating, anti-inflammatory, anti-apoptotic, anti-thrombotic, natriuretic, and cardioprotective effects. epoxyeicosatrienoic acids 82-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-38 20093140-4 2010 CYP epoxygenases such as CYP1A2, CYP2C, and CYP2J2 metabolize arachidonic acid to epoxyeicosatrienoic acids (EETs) which generally possess vasodilating, anti-inflammatory, anti-apoptotic, anti-thrombotic, natriuretic, and cardioprotective effects. eets 109-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-38 19717402-1 2010 AIMS: A CYP2C9-dependent endothelium-derived hyperpolarizing factor (EDHF) controls blood flow in many microvascular beds of various species by targeting vascular smooth muscle potassium channels. 1-hexadecyl-2-acetyl-glycero-3-phosphocholine 61-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 19717402-1 2010 AIMS: A CYP2C9-dependent endothelium-derived hyperpolarizing factor (EDHF) controls blood flow in many microvascular beds of various species by targeting vascular smooth muscle potassium channels. edhf 69-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 19717402-6 2010 Local superfusion of the CYP2C9 inhibitor sulfaphenazole, at doses known to block EDHF-dependent dilations, significantly augmented PVWI, as did inhibition of NO synthase. Sulfaphenazole 42-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19717402-6 2010 Local superfusion of the CYP2C9 inhibitor sulfaphenazole, at doses known to block EDHF-dependent dilations, significantly augmented PVWI, as did inhibition of NO synthase. edhf 82-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19717402-9 2010 The prothrombotic effects of CYP2C9 inhibition in vivo were reversed by exogenous superfusion with 11,12-epoxyeicosatrienoic acids. 11,12-epoxy-5,8,14-eicosatrienoic acid 99-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19717402-12 2010 CONCLUSION: The arteriolar endothelium in vivo continuously releases a CYP2C9-derived EDHF. edhf 86-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 19925783-0 2010 AFM study of the interaction of cytochrome P450 2C9 with phospholipid bilayers. Phospholipids 57-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 19925783-2 2010 In this study, atomic force microscopy was applied to characterise the association of CYP2C9 to dimyristoylphosphatidylcholine (DMPC) supported phospholipid bilayers. Dimyristoylphosphatidylcholine 96-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 19925783-2 2010 In this study, atomic force microscopy was applied to characterise the association of CYP2C9 to dimyristoylphosphatidylcholine (DMPC) supported phospholipid bilayers. Dimyristoylphosphatidylcholine 128-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 19925783-2 2010 In this study, atomic force microscopy was applied to characterise the association of CYP2C9 to dimyristoylphosphatidylcholine (DMPC) supported phospholipid bilayers. Phospholipids 144-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 19925783-3 2010 CYP2C9 was found to exclusively localise in the gel domains of partially melted DMPC bilayers. Dimyristoylphosphatidylcholine 80-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19925783-4 2010 Despite lacking the N-terminus transmembrane spanning domain, the CYP2C9 protein appeared to partially embed into the membrane bilayer, as evidenced by an increase in melting temperature of surrounding phospholipids. Phospholipids 202-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 19925783-5 2010 Reversible binding of CYP2C9 via an engineered His tag to a phospholipid bilayer was facilitated using nickel-chelating lipids, presenting potential applications for biosensor technologies. Histidine 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 19925783-5 2010 Reversible binding of CYP2C9 via an engineered His tag to a phospholipid bilayer was facilitated using nickel-chelating lipids, presenting potential applications for biosensor technologies. Phospholipids 60-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 19925783-5 2010 Reversible binding of CYP2C9 via an engineered His tag to a phospholipid bilayer was facilitated using nickel-chelating lipids, presenting potential applications for biosensor technologies. Nickel 103-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20233183-0 2010 Time-dependent inhibition (TDI) of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Noscapine 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 20233183-0 2010 Time-dependent inhibition (TDI) of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 20233183-6 2010 Kinetic analysis showed that inhibition of CYP2C9 by noscapine was best fit to a noncompetitive type with K(i) value of 8.8 microm, while inhibition of CYP3A4 by noscapine was best fit to a competitive manner with K(i) value of 5.2 microm. Noscapine 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20233183-7 2010 Noscapine also exhibited TDI to CYP3A4 and CYP2C9. Noscapine 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20233183-10 2010 CONCLUSIONS: TDI of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Noscapine 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 20233183-10 2010 CONCLUSIONS: TDI of CYP3A4 and CYP2C9 by noscapine potentially explains clinical noscapine-warfarin interaction. Noscapine 81-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 19855097-0 2010 CYP2C9*1B promoter polymorphisms, in linkage with CYP2C19*2, affect phenytoin autoinduction of clearance and maintenance dose. Phenytoin 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19567378-0 2010 Pharmacogenetic impact of VKORC1 and CYP2C9 allelic variants on warfarin dose requirements in a hispanic population isolate. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19567378-9 2010 A stepwise multivariate linear regression analysis showed that 38.2% of the warfarin dose response variance was accounted for by a model involving age (20.9%), VKORC1-1639G/A (11.3%), and CYP2C9*1, *2, and *3 variants (7.1%). Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 20022989-7 2010 The role of cytochrome P450 epoxygenase (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) was assessed by repeating acetylcholine under intraarterial sulfaphenazole. endothelium-derived hyperpolarizing factor 58-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 20022989-7 2010 The role of cytochrome P450 epoxygenase (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) was assessed by repeating acetylcholine under intraarterial sulfaphenazole. edhf 102-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 20022989-7 2010 The role of cytochrome P450 epoxygenase (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) was assessed by repeating acetylcholine under intraarterial sulfaphenazole. Acetylcholine 134-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 20022989-7 2010 The role of cytochrome P450 epoxygenase (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) was assessed by repeating acetylcholine under intraarterial sulfaphenazole. Sulfaphenazole 168-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 20022989-15 2010 A compensatory CYP 2C9-derived EDHF pathway is activated to sustain endothelium-dependent vasodilation. edhf 31-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-22 19941044-10 2010 The presence of a single *2 or *3 CYP2C9 allele reduced mean [SE (standard error)] S-warfarin clearance by 35% from 0.276 (0.04) to 0.180 (0.11) l/h. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19941044-13 2010 Steady state simulations showed that whilst CYP2C9 polymorphisms affect the PK of warfarin, VKORC1 haplotypes may be better predictors of warfarin response. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 20121287-10 2010 CONCLUSION: Genotyping of four SNPs for VKORC1 and CYP2C9 polymorphisms is useful in predicting a high probability of the occurrence of DAH in patients receiving oral anticoagulants. dah 136-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 20206792-2 2010 OBJECTIVE: The aim of this study was to determine whether folic acid supplementation increases the dosage requirement of the CYP2C9 substrate warfarin, and the formation clearance of the CYP2C9-mediated product, (S)-7-hydroxywarfarin. Folic Acid 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 20206792-2 2010 OBJECTIVE: The aim of this study was to determine whether folic acid supplementation increases the dosage requirement of the CYP2C9 substrate warfarin, and the formation clearance of the CYP2C9-mediated product, (S)-7-hydroxywarfarin. 7-hydroxywarfarin 212-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 19847408-0 2010 The effects of CYP2C9 and CYP2C19 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of glipizide in Chinese subjects. Glipizide 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 19847408-1 2010 OBJECTIVE: To study the effects of CYP2C9 and CYP2C19 genetic polymorphisms on the pharmacokinetics and pharmacodynamics of glipizide. Glipizide 124-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 19847408-8 2010 CONCLUSIONS: These results suggest that CYP2C9 polymorphism significantly influences the pharmacokinetics and pharmacodynamics of glipizide, which needs to be considered in clinical practice. Glipizide 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). Arachidonic Acid 90-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-61 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). epoxyeicosatrienoic acids 110-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-61 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). eets 137-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-61 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dihydroxyeicosatrienoic acids 176-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-61 19891972-1 2010 The cytochrome P450 (CYP) epoxygenase enzymes CYP2J and CYP2C catalyze the epoxidation of arachidonic acid to epoxyeicosatrienoic acids (EETs), which are rapidly hydrolyzed to dihydroxyeicosatrienoic acids (DHETs) by soluble epoxide hydrolase (sEH). dhets 207-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-61 20121287-2 2010 OBJECTIVE: We hypothesized that in patients treated with coumarins, alveolar hemorrhage is associated with vitamin K epoxide reductase (VKORC1) and cytochrome P450 (CYP) 2C9 (CYP2C9) variant alleles. Coumarins 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-173 20121287-2 2010 OBJECTIVE: We hypothesized that in patients treated with coumarins, alveolar hemorrhage is associated with vitamin K epoxide reductase (VKORC1) and cytochrome P450 (CYP) 2C9 (CYP2C9) variant alleles. Coumarins 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 19855097-1 2010 The commonly prescribed antiepileptic drug phenytoin has a narrow therapeutic range and wide interindividual variability in clearance explained in part by CYP2C9 and CYP2C19 coding variants. Phenytoin 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 19855097-2 2010 After finding a paradoxically low urinary phenytoin metabolite (S)/(R) ratio in subjects receiving phenytoin maintenance therapy with a CYP2C9*1/*1 and CYP2C19*1/*2 genotype, we hypothesized that CYP2C9 regulatory polymorphisms (rPMs), G-3089A and -2663delTG, in linkage disequilibrium with CYP2C19*2 were responsible. Phenytoin 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 19855097-2 2010 After finding a paradoxically low urinary phenytoin metabolite (S)/(R) ratio in subjects receiving phenytoin maintenance therapy with a CYP2C9*1/*1 and CYP2C19*1/*2 genotype, we hypothesized that CYP2C9 regulatory polymorphisms (rPMs), G-3089A and -2663delTG, in linkage disequilibrium with CYP2C19*2 were responsible. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 19855097-4 2010 We hypothesized the rPMs affected CYP2C9 induction by phenytoin, a pregnane X receptor (PXR), and constitutive androstane receptor (CAR) activator. rpms 20-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19855097-5 2010 Transfection studies showed that CYP2C9 reporters with wild-type versus variant alleles had similar basal activity but significantly greater phenytoin induction by cotransfected PXR, CAR, and Nrf2 and less Yin Yang 1 transcription factor repression. Phenytoin 141-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 19855097-6 2010 Phenytoin induction of CYP2C9 was greater in human hepatocytes with the CYP2C9 wild type versus variant haplotype. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 19855097-6 2010 Phenytoin induction of CYP2C9 was greater in human hepatocytes with the CYP2C9 wild type versus variant haplotype. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. rpms 18-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. rpms 18-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. rpms 18-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. Phenytoin 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. Phenytoin 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. Phenytoin 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19855097-7 2010 Therefore, CYP2C9 rPMs affect phenytoin-dependent induction of CYP2C9 and phenytoin metabolism in humans, with an effect size comparable with that for CYP2C9*2 and 2C9*3. Phenytoin 74-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 19686083-5 2010 Although the polymorphisms in CYP2C9 and VKORC1 explain a significant proportion of the interindividual variability in warfarin dose requirement, currently available evidence based on a few small studies relating to the use of pharmacogenetics-guided dosing in the initiation of warfarin therapy has not shown improved outcomes in either safety or efficacy of therapy. Warfarin 279-287 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 21215088-4 2010 One hundred and one patients with stable anticoagulation with warfarin under a target international normalized ratio (INR) of 2.0 to 3.0 were enrolled for studying the relationship between the CYP2C9 and VKORC1 gene polymorphism and the warfarin maintaining dosage. Warfarin 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 19686083-3 2010 Single nucleotide polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR) genes have been shown to have a significant effect on warfarin dose requirement. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 19903527-2 2010 Additionally, the influence of optimized GRDDS on the bioavailability of Losartan and the formation extent of active metabolite E3174 by CYP2C9 polymorphism was investigated. losartan carboxylic acid 128-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 19903527-9 2010 The lower bioavailability of Losartan in the CYP2C9*1/*1 subjects than CYP2C9*1/*3 subjects was found and could be due to the variety of enzymatic activity. Losartan 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 19903527-9 2010 The lower bioavailability of Losartan in the CYP2C9*1/*1 subjects than CYP2C9*1/*3 subjects was found and could be due to the variety of enzymatic activity. Losartan 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 19686083-3 2010 Single nucleotide polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR) genes have been shown to have a significant effect on warfarin dose requirement. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 19686083-5 2010 Although the polymorphisms in CYP2C9 and VKORC1 explain a significant proportion of the interindividual variability in warfarin dose requirement, currently available evidence based on a few small studies relating to the use of pharmacogenetics-guided dosing in the initiation of warfarin therapy has not shown improved outcomes in either safety or efficacy of therapy. Warfarin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 20834094-3 2010 Etravirine is a weak inducer of cytochrome P450 (CYP)3A and a weak inhibitor of CYP2C9/CYP2C19 and P-glycoprotein, and although etravirine is metabolized by the CYP enzyme system, the extent of clinically relevant interactions with other antiretrovirals is limited. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 19794411-4 2010 In addition to age, genetic variants of VKORC1, CYP2C9, CYP4F2, and EPHX1 were found to be significant predictor variables for the maintenance dose of warfarin, explaining 26.6% of dose variability. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 20671989-5 2010 The exact mechanism of this interaction is unknown but may be related to downregulation of cytochrome P450 2C9 by capecitabine or its metabolites. Capecitabine 114-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-110 19794411-0 2010 Genetic factors (VKORC1, CYP2C9, EPHX1, and CYP4F2) are predictor variables for warfarin response in very elderly, frail inpatients. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19794412-0 2010 Loss-of-function CYP2C9 variants improve therapeutic response to sulfonylureas in type 2 diabetes: a Go-DARTS study. Sulfonylurea Compounds 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19794412-1 2010 Sulfonylureas are metabolized mainly by the cytochrome p450 2C9 (CYP2C9) enzyme. Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-63 19794412-1 2010 Sulfonylureas are metabolized mainly by the cytochrome p450 2C9 (CYP2C9) enzyme. Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19794412-7 2010 In conclusion, CYP2C9 loss-of-function alleles are associated with greater response to sulfonylureas and decreased failure of therapy consistent with the pharmacokinetic role of CYP2C9. Sulfonylurea Compounds 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 20205664-1 2010 The identification of the genes encoding CYP2C9, the principal metabolizing enzyme of the coumarins, and VKORC1, the molecular target for coumarins, has strongly stimulated the research on pharmacogenetics of vitamin K antagonists, also designated as coumarins. Coumarins 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 20171411-1 2010 BACKGROUND: Bosentan is an endothelin-receptor antagonist that reportedly induces both cytochrome P450 (CYP) 3A4 and CYP2C9 enzymes, which are also involved in warfarin metabolism. Bosentan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 20171411-1 2010 BACKGROUND: Bosentan is an endothelin-receptor antagonist that reportedly induces both cytochrome P450 (CYP) 3A4 and CYP2C9 enzymes, which are also involved in warfarin metabolism. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 20171411-7 2010 CONCLUSIONS: Bosentan has CYP3A4- and CYP2C9-inducing properties and is therefore likely to cause decreased concentrations of warfarin. Bosentan 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 20877134-0 2010 Flavonoids diosmetin and hesperetin are potent inhibitors of cytochrome P450 2C9-mediated drug metabolism in vitro. Flavonoids 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-80 20877134-0 2010 Flavonoids diosmetin and hesperetin are potent inhibitors of cytochrome P450 2C9-mediated drug metabolism in vitro. diosmetin 11-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-80 20877134-0 2010 Flavonoids diosmetin and hesperetin are potent inhibitors of cytochrome P450 2C9-mediated drug metabolism in vitro. hesperetin 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-80 20877134-1 2010 The aim of this study was to examine in vitro, by means of kinetic analysis and molecular docking simulations, the effects of the flavone diosmetin and its flavanone analog hesperetin on CYP (cytochrome P450) 2C9-mediated drug metabolism. flavone 130-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-212 20877134-1 2010 The aim of this study was to examine in vitro, by means of kinetic analysis and molecular docking simulations, the effects of the flavone diosmetin and its flavanone analog hesperetin on CYP (cytochrome P450) 2C9-mediated drug metabolism. diosmetin 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-212 20877134-1 2010 The aim of this study was to examine in vitro, by means of kinetic analysis and molecular docking simulations, the effects of the flavone diosmetin and its flavanone analog hesperetin on CYP (cytochrome P450) 2C9-mediated drug metabolism. flavanone 156-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-212 20877134-2 2010 To this purpose, the conversion of diclofenac to 4"-hydroxydiclofenac by human liver microsomes was used as a model assay for assessing the CYP2C9 inhibitory activity of these two flavonoids. 4'-hydroxydiclofenac 49-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 20877134-2 2010 To this purpose, the conversion of diclofenac to 4"-hydroxydiclofenac by human liver microsomes was used as a model assay for assessing the CYP2C9 inhibitory activity of these two flavonoids. Flavonoids 180-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 20877134-6 2010 The results of molecular docking simulations were consistent with those of kinetic analysis, since they showed that the putative binding sites of both diosmetin and hesperetin coincided with the CYP2C9 substrate binding site. diosmetin 151-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 20877134-6 2010 The results of molecular docking simulations were consistent with those of kinetic analysis, since they showed that the putative binding sites of both diosmetin and hesperetin coincided with the CYP2C9 substrate binding site. hesperetin 165-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. diosmetin 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. diosmetin 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. hesperetin 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. hesperetin 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 20930419-0 2010 Pharmacokinetic and pharmacodynamic variation associated with VKORC1 and CYP2C9 polymorphisms in Thai patients taking warfarin. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 20930419-1 2010 We investigated the influence of genetic polymorphisms of CYP2C9 and VKORC1 genotypes on the pharmacokinetics and pharmacodynamics of warfarin and established an equation for predicting the maintenance dose of warfarin in the Thai population using genetic and non-genetic factors. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 20930419-1 2010 We investigated the influence of genetic polymorphisms of CYP2C9 and VKORC1 genotypes on the pharmacokinetics and pharmacodynamics of warfarin and established an equation for predicting the maintenance dose of warfarin in the Thai population using genetic and non-genetic factors. Warfarin 210-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 20930419-4 2010 CYP2C9 polymorphisms affect warfarin metabolism as shown by a significant difference in warfarin clearance, whereas VKORC1 genotypes cause a significant difference in warfarin sensitivity index (INR:Cp). Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20930419-4 2010 CYP2C9 polymorphisms affect warfarin metabolism as shown by a significant difference in warfarin clearance, whereas VKORC1 genotypes cause a significant difference in warfarin sensitivity index (INR:Cp). Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20930419-4 2010 CYP2C9 polymorphisms affect warfarin metabolism as shown by a significant difference in warfarin clearance, whereas VKORC1 genotypes cause a significant difference in warfarin sensitivity index (INR:Cp). Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20930419-5 2010 The mean weekly warfarin dose was significantly different among different VKORC1 and CYP2C9 genotypes. Warfarin 16-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 20930419-6 2010 Patients with the VKORC1 BB haplotype and CYP2C9*1/*1 required about twice the warfarin dose compared to those with the VKORC1 AA haplotype and CYP2C9*1/*1. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 20930419-7 2010 Using stepwise multiple linear regression, clinical factors (age and weight) and genetic factors (CYP2C9 and VKORC1) could explain about 53.8% of the variance of the warfarin maintenance dose. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 20930419-8 2010 CYP2C9 and VKORC1 genotypes played an important role in the inter-individual variation in warfarin maintenance dose in a Thai population. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20877134-7 2010 The demonstration that diosmetin and hesperetin inhibit CYP2C9-mediated diclofenac metabolism at low micromolar concentrations is of potential clinical relevance because CYP2C9 is responsible for the biotransformation of various therapeutically important drugs that have narrow therapeutic indexes. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 20962433-1 2010 Benzbromarone is a uricosuric drug and has been shown to be metabolized predominantly by cytochrome P450(CYP)2C9 in vitro findings. Benzbromarone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-112 20962433-2 2010 This study aims to investigate the influence of the CYP2C9 genotype on plasma levels of benzbromarone and 6-hydroxybenzbromarone, as well as uric acid lowering effects. Benzbromarone 88-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 20962433-2 2010 This study aims to investigate the influence of the CYP2C9 genotype on plasma levels of benzbromarone and 6-hydroxybenzbromarone, as well as uric acid lowering effects. 6-hydroxybenzbromarone 106-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 20962433-4 2010 The oral clearance of benzbromarone in the CYP2C9*1/*1 genotype and CYP2C9*1/*3 genotype was 58.8+-25.2 L/hr/kg (mean+-SD) and 51.3+-7.9 L/hr/kg, respectively, whereas 8.58 L/hr/kg in the CYP2C9*3/*3 genotype. Benzbromarone 22-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20962433-5 2010 The metabolic ratio (6-hydroxybenzbromarone/benzbromarone) in urine was 38.6+-10.7 in the CYP2C9*1/*1 genotype, 35.4+-12.4 in the CYP2C9*1/*3 genotype and 12.9 in the CYP2C9*3/*3 genotype. 6-hydroxybenzbromarone 21-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 20962433-7 2010 These results suggest a critical role for CYP2C9 in the metabolism of benzbromarone in humans and a possible risk of toxicity in the CYP2C9*3 homozygote by lowering clearance of the drug. Benzbromarone 70-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 20962433-8 2010 Further studies are required to assess the clinical impact of CYP2C9 on the metabolism of benzbromarone. Benzbromarone 90-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 20205664-1 2010 The identification of the genes encoding CYP2C9, the principal metabolizing enzyme of the coumarins, and VKORC1, the molecular target for coumarins, has strongly stimulated the research on pharmacogenetics of vitamin K antagonists, also designated as coumarins. Coumarins 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 20205664-1 2010 The identification of the genes encoding CYP2C9, the principal metabolizing enzyme of the coumarins, and VKORC1, the molecular target for coumarins, has strongly stimulated the research on pharmacogenetics of vitamin K antagonists, also designated as coumarins. Vitamin K 209-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 20205664-1 2010 The identification of the genes encoding CYP2C9, the principal metabolizing enzyme of the coumarins, and VKORC1, the molecular target for coumarins, has strongly stimulated the research on pharmacogenetics of vitamin K antagonists, also designated as coumarins. Coumarins 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 20205664-2 2010 From 1999 to 2004 a number of observational studies firmly established associations between being carrier of the CYP2C9*2 and especially the CYP2C9*3 allele and reduced coumarin dose requirements and increased risks of overanticoagulation and even major bleeding compared to CYP2C9 wild type patients. coumarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 20205664-2 2010 From 1999 to 2004 a number of observational studies firmly established associations between being carrier of the CYP2C9*2 and especially the CYP2C9*3 allele and reduced coumarin dose requirements and increased risks of overanticoagulation and even major bleeding compared to CYP2C9 wild type patients. coumarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 20205664-2 2010 From 1999 to 2004 a number of observational studies firmly established associations between being carrier of the CYP2C9*2 and especially the CYP2C9*3 allele and reduced coumarin dose requirements and increased risks of overanticoagulation and even major bleeding compared to CYP2C9 wild type patients. coumarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 22134997-3 2010 Polymorphisms of two genes encoding enzymes involved in vitamin K and/or VKA metabolism such as vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 isoform (CYP2C9) play a key role in this variance. Vitamin K 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-174 21152245-9 2010 Strong correlations between allelic variation in the tamoxifen pathway at CYP1A1-CYP3A4, CYP3A4-CYP2C9, and CYP2C9-SULT1A2, in addition to CYP2D6 and its adjacent genes, were seen in the placebo-arm but not the tamoxifen-arm. Tamoxifen 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 19883715-3 2010 In this study, we systematically evaluated the inhibitory effects of 60 polyphenols and related compounds on human cytochrome P450 (CYP) 3A4 and CYP2C9 activity by in vitro assay to investigate whether some polyphenols induce drug interactions. Polyphenols 72-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 19883715-5 2010 Three coumarins and 12 flavonoids significantly suppressed CYP3A4 or CYP2C9 activities. Coumarins 6-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 19883715-5 2010 Three coumarins and 12 flavonoids significantly suppressed CYP3A4 or CYP2C9 activities. Flavonoids 23-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 19883715-6 2010 Lineweaver-Burk plot analysis indicated that apigenin and its dimer amentoflavone and imperatorin displayed a mixed type of inhibition on CYP3A4 or CYP2C9. amentoflavone 68-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 19883715-7 2010 Among the inhibitors, amentoflavone was the most potent inhibitor of CYP3A4 and CYP2C9 activities with IC(50) values of 0.07 and 0.03 microM, respectively. amentoflavone 22-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 19883715-8 2010 The K(i) value of amentoflavone was significantly lower than that of the CYP2C9 inhibition positive control sulfaphenazole. amentoflavone 18-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19883715-8 2010 The K(i) value of amentoflavone was significantly lower than that of the CYP2C9 inhibition positive control sulfaphenazole. Sulfaphenazole 108-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 20393255-2 2010 Voriconazole, a CYP2C9 inhibitor, has many potential drug interactions. Voriconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 19874474-0 2010 Ability of VKORC1 and CYP2C9 to predict therapeutic warfarin dose during the initial weeks of therapy. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 19874474-1 2010 BACKGROUND: CYP2C9 and VKORC1 genotypes predict therapeutic warfarin dose at initiation of therapy; however, the predictive ability of genetic information after a week or longer is unknown. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 22134997-3 2010 Polymorphisms of two genes encoding enzymes involved in vitamin K and/or VKA metabolism such as vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 isoform (CYP2C9) play a key role in this variance. Vitamin K 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 20014877-1 2010 BACKGROUND: In 2007, the US FDA added information about pharmacogenomics to the warfarin label based on the influence of the CYP2C9 and VKORC1 genes on anticoagulation-related outcomes. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 20617424-3 2010 Significant findings have been documented showing the effect of certain genetic variations (e.g., in CYP2C9 and VKORC1) on the dose response to warfarin. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 19934793-0 2010 Besides CYP2C19*2, the variant allele CYP2C9*3 is associated with higher on-clopidogrel platelet reactivity in patients on dual antiplatelet therapy undergoing elective coronary stent implantation. Clopidogrel 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 20017677-1 2010 AIMS: VKORC1 and CYP2C9 are important genetic factors affecting warfarin dose requirement. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19934793-10 2010 CONCLUSION: Besides CYP2C19*2, the variant allele CYP2C9*3 plays an important role in the response to clopidogrel in patients on dual antiplatelet therapy undergoing coronary stenting. Clopidogrel 102-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19614891-0 2009 The role of human CYP2C8 and CYP2C9 variants in pioglitazone metabolism in vitro. Pioglitazone 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19952982-2 2010 The goal of this study was to assess the effect of maternal genotype of functional polymorphisms in two genes involved in phenytoin metabolism, CYP2C9 (R144C, I395L) and EPHX1 (Y113H, H139R), on the presence of major craniofacial abnormalities (CFAs) in the child. Phenytoin 122-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 20597268-15 2010 The determination of CYP2C9 and VCORC1 genotypes in patients with APS before warfarin use allows excessive hypocoagulation and related hemorrhages to be avoided. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 19614891-6 2009 Mean intrinsic clearance of pioglitazone to the metabolite M-IV was highest for CYP2C8 and CYP1A2 with 58 pmol M-IV/min/nmol CYP P450/microM pioglitazone each, 53 for CYP2D6*1, 40 for CYP2C19*1, and 34 for CYP2C9*2, respectively. Pioglitazone 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 19614891-3 2009 Pioglitazone (2-400 microM) was incubated with isolated cytochrome P450 enzymes or human liver microsomes, some of them carrying either the CYP2C8*3/*3 genotype (and also the CYP2C9*2/*2 genotype) or the CYP2C8*1/*1 genotype (five samples each). Pioglitazone 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 20116625-3 2009 ETR acts as an inducer of CYP3A4 and inhibitor of CYP2C9 and CYP2C19. etravirine 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19756559-0 2009 Effects of CYP2C19 and CYP2C9 genotypes on pharmacokinetic variability of valproic acid in Chinese epileptic patients: nonlinear mixed-effect modeling. Valproic Acid 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 19756559-1 2009 PURPOSE: To evaluate the effects of CYP2C19 and CYP2C9 genotypes on the pharmacokinetic variability of valproic acid (VPA) in epileptic patients using a population pharmacokinetic (PPK) approach. Valproic Acid 103-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 19958586-1 2009 OBJECTIVES: It has been reported that hepatic cytochrome P450 (CYP)2C9 and CYP3A4 are responsible for the metabolism of sildenafil and formation of its metabolite, N-desmethylsildenafil, in humans. Sildenafil Citrate 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-70 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dexamethasone 262-265 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20167002-6 2009 The results of the analysis suggest that in addition to the potent function of GR and the further involvement of PXR and CAR activated by DXM or DHEA, an additional factor might play a role in CYP2C9 regulation by DHEA. Dehydroepiandrosterone 214-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 20167002-7 2009 The novel potential candidate for DHEA action in CYP2C9 induction is likely to be the estrogen receptor. Dehydroepiandrosterone 34-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 20167002-8 2009 Additionally, the balance of DHEA sulphation-desulphation processes should also be considered in any description of DHEA-induced CYP2C9 profiles. Dehydroepiandrosterone 116-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 20167000-9 2009 Currently, there are three X-ray structures of the human CYP2C9 in Protein Database (PDB): one ligand-free protein (1OG2), and two in complex with S-warfarin (1OG5) or flurbiprofen (1R9O). Sulfur 147-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 20167000-9 2009 Currently, there are three X-ray structures of the human CYP2C9 in Protein Database (PDB): one ligand-free protein (1OG2), and two in complex with S-warfarin (1OG5) or flurbiprofen (1R9O). Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 20167000-9 2009 Currently, there are three X-ray structures of the human CYP2C9 in Protein Database (PDB): one ligand-free protein (1OG2), and two in complex with S-warfarin (1OG5) or flurbiprofen (1R9O). Flurbiprofen 168-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 20167000-13 2009 In the structures of CYP2C9 without ligand bound or with bound S-warfarin, residues 101-106 in the B-C loop form helix B". Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 20167000-17 2009 CYP2C9 is activated by dapsone and its analogues and R-lansoprazole in a stereo-specific and substrate-dependent manner, probably through binding to the active site and inducing positive cooperativity. Dapsone 23-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167000-17 2009 CYP2C9 is activated by dapsone and its analogues and R-lansoprazole in a stereo-specific and substrate-dependent manner, probably through binding to the active site and inducing positive cooperativity. Dexlansoprazole 53-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167001-5 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Steroids 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167001-5 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Melatonin 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167001-5 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Retinoids 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167001-5 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Arachidonic Acid 118-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20167001-6 2009 Typical substrates of CYP2C9 such as celecoxib, ibuprofen, flurbiprofen, and diclofenac are relatively small, lipophilic and contain acidic groupings with pK(a) values in the range 3.8-8.1 which will be ionized at physiological pH. Celecoxib 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20167001-6 2009 Typical substrates of CYP2C9 such as celecoxib, ibuprofen, flurbiprofen, and diclofenac are relatively small, lipophilic and contain acidic groupings with pK(a) values in the range 3.8-8.1 which will be ionized at physiological pH. Ibuprofen 48-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20167001-6 2009 Typical substrates of CYP2C9 such as celecoxib, ibuprofen, flurbiprofen, and diclofenac are relatively small, lipophilic and contain acidic groupings with pK(a) values in the range 3.8-8.1 which will be ionized at physiological pH. Flurbiprofen 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20167001-6 2009 Typical substrates of CYP2C9 such as celecoxib, ibuprofen, flurbiprofen, and diclofenac are relatively small, lipophilic and contain acidic groupings with pK(a) values in the range 3.8-8.1 which will be ionized at physiological pH. Diclofenac 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 20167001-7 2009 The carboxylate groups of tienilic acid and diclofenac have been shown to be responsible for substrate preference and orientation in the active site of CYP2C9. carboxylate 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 20167001-7 2009 The carboxylate groups of tienilic acid and diclofenac have been shown to be responsible for substrate preference and orientation in the active site of CYP2C9. Ticrynafen 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 20167001-7 2009 The carboxylate groups of tienilic acid and diclofenac have been shown to be responsible for substrate preference and orientation in the active site of CYP2C9. Diclofenac 44-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 20167001-12 2009 Pharmacophore models have revealed that hydrogen bonding, ion-pair interactions, and probably hydrophobic interactions play a major role in determining the substrate specificity and inhibitor selectivity of CYP2C9. Hydrogen 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Steroids 158-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Steroids 158-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dehydroepiandrosterone 184-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dehydroepiandrosterone 184-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dehydroepiandrosterone 208-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dehydroepiandrosterone 208-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dexamethasone 247-260 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dexamethasone 247-260 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 20167002-5 2009 As an example, we present modelling and simulation approaches of the CYP2C9 gene expression in human hepatocytes treated with well-known CYP2C9 inducers: the steroid hormone precursor dehydroepiandrosterone (DHEA) and the synthetic glucocorticoid dexamethasone (DXM). Dexamethasone 262-265 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 20043560-3 2009 Vitamin K epoxide reductase (VKORC1) and cytochrome P450 2C9 (CYP2C9) enzyme conjointly determine the warfarin maintenance dose. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-60 20043560-3 2009 Vitamin K epoxide reductase (VKORC1) and cytochrome P450 2C9 (CYP2C9) enzyme conjointly determine the warfarin maintenance dose. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19958586-1 2009 OBJECTIVES: It has been reported that hepatic cytochrome P450 (CYP)2C9 and CYP3A4 are responsible for the metabolism of sildenafil and formation of its metabolite, N-desmethylsildenafil, in humans. Sildenafil Citrate 164-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-70 19955245-4 2009 For the 189 Mayo Clinic patients initiating warfarin therapy to achieve a target international normalized ratio (INR) in the range of 2.0 to 3.5, we analyzed the CYP2C9 (cytochrome P450 2C9) and VKORC1 (vitamin K epoxide reductase complex, subunit 1) genetic loci to study the relationship among the initial warfarin dose, steady-state dose, time to achieve steady-state dose, variations in INR, and allelic variance. Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19955245-4 2009 For the 189 Mayo Clinic patients initiating warfarin therapy to achieve a target international normalized ratio (INR) in the range of 2.0 to 3.5, we analyzed the CYP2C9 (cytochrome P450 2C9) and VKORC1 (vitamin K epoxide reductase complex, subunit 1) genetic loci to study the relationship among the initial warfarin dose, steady-state dose, time to achieve steady-state dose, variations in INR, and allelic variance. Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-189 19955245-9 2009 Polymorphisms in CYP2C9 and VKORC1 have a considerable effect on warfarin dose in white people. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19958090-2 2009 Polymorphisms in CYP2C9 and VKORC1 have been shown to be associated with warfarin dose requirements. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19955245-10 2009 A correlation between steady-state warfarin dose and allelic variants of CYP2C9 and VKORC1 has been demonstrated by many previous reports and is reconfirmed in this report. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19881396-1 2009 OBJECTIVE: Variants of two genes, CYP2C9 and VKORC1, explain approximately one third of variability in warfarin maintenance dose requirements. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19881396-10 2009 CONCLUSION: Our initial results imply that pharmacogenomic-guided warfarin dosing may be more useful in settings with less intensive patient follow-up, and when adjustments are made for slower therapeutic response in patients with a CYP2C9 variant. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 22478935-11 2009 Drug-drug interactions with other substrates of CYP2C9, CYP2C19, and CYP3A4 can also alter voriconazole concentrations. Voriconazole 91-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 19843839-8 2009 Although the exact mechanism of the interaction is unknown, one theory is that tamoxifen inhibits CYP2C9, which metabolizes the S-isomer of warfarin. Tamoxifen 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 19843839-8 2009 Although the exact mechanism of the interaction is unknown, one theory is that tamoxifen inhibits CYP2C9, which metabolizes the S-isomer of warfarin. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 19663817-1 2009 Fluvastatin has been considered to be metabolised to 5-hydroxy fluvastatin (M-2), 6-hydroxy fluvastatin (M-3) and N-desisopropyl fluvastatin (M-5) in human liver microsomes by primarily CYP2C9. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 20214591-4 2009 Among the enzymes involved in aspirin biodisposition a major role is played by the enzymes UDP-glucuronosyltransferase UGT1A6, cytochrome P450 CYP2C9 and the xenobiotic/medium chain fatty acid:CoA ligase ACSM2, although other UGTs and ACSMs enzymes may significantly contribute to aspirin metabolism. Aspirin 30-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 20214591-4 2009 Among the enzymes involved in aspirin biodisposition a major role is played by the enzymes UDP-glucuronosyltransferase UGT1A6, cytochrome P450 CYP2C9 and the xenobiotic/medium chain fatty acid:CoA ligase ACSM2, although other UGTs and ACSMs enzymes may significantly contribute to aspirin metabolism. Aspirin 281-288 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 20214591-7 2009 Major polymorphisms related to aspirin biodisposition are rs2070959, rs1105879 and rs6759892 for the UGT1A6 gene, rs1133607 for the ACSM2 gene, and rs1799853, rs1057910, rs28371686, rs9332131 and rs28371685 for the CYP2C9 gene. Aspirin 31-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 215-221 19651758-8 2009 In contrast, activity of CYP2A6 (p = 0.001) and CYP2C9 (diclofenac, p = 0.0001; tolbutamide, p = 0.004) was significantly increased with NAFLD progression. Tolbutamide 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 19582440-3 2009 PCR-based methods were performed to analyze genetic polymorphisms in the genes pharmacokinetically and pharmacodynamically related to warfarin reactions, including cytochrome P450 (CYP) 2C9, vitamin K epoxide reductase complex subunit 1 (VKORC1), gamma-glutamyl carboxylase (GGCX) and factor VII (FVII). Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-189 19582440-4 2009 RESULTS: The presence of CYP2C9*3 and VKORC1-1639G>A had a significant impact on the mean maintenance dose of warfarin (CYP2C9*1/*1 2.74 +/- 1.24 mg/day vs. *1/*3 and *3/*3 1.56 +/- 0.85 mg/day, P = 0.009; VKORC1-1639AA 2.42 +/- 0.95 mg/day vs. GA 3.71 +/- 1.43 mg/day vs. GG 7.25 +/- 0.35 mg/day, P < 0.001). Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19582440-4 2009 RESULTS: The presence of CYP2C9*3 and VKORC1-1639G>A had a significant impact on the mean maintenance dose of warfarin (CYP2C9*1/*1 2.74 +/- 1.24 mg/day vs. *1/*3 and *3/*3 1.56 +/- 0.85 mg/day, P = 0.009; VKORC1-1639AA 2.42 +/- 0.95 mg/day vs. GA 3.71 +/- 1.43 mg/day vs. GG 7.25 +/- 0.35 mg/day, P < 0.001). Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 19582440-5 2009 In the multiple linear regression model, the combination of age, body surface area, and genotypes of CYP2C9*3 and VKORC1-1639G>A explained 54.8% of the variance in warfarin dose requirements. Warfarin 167-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 19582440-6 2009 CONCLUSIONS: The influences of CYP2C9*3 and VKORC1-1639G>A on the maintenance dose of warfarin were well-defined in Japanese patients, while polymorphisms of GGCX and FVII did not affect it. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 19651758-8 2009 In contrast, activity of CYP2A6 (p = 0.001) and CYP2C9 (diclofenac, p = 0.0001; tolbutamide, p = 0.004) was significantly increased with NAFLD progression. Diclofenac 56-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 19891554-0 2009 Presence of CYP2C9*3 allele increases risk for hypoglycemia in Type 2 diabetic patients treated with sulfonylureas. Sulfonylurea Compounds 101-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 19891554-2 2009 Impaired metabolism of sulfonylureas due to gene polymorphisms in the metabolic enzyme CYP2C9 might lead to hypoglycemia. Sulfonylurea Compounds 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. Sulfonylurea Compounds 178-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. Sulfonylurea Compounds 178-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. Sulfonylurea Compounds 178-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. glimepiride 192-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. glimepiride 192-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. glimepiride 192-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19891554-3 2009 In the present study we explored the association of the CYP2C9 variant alleles CYP2C9*2 and CYP2C9*3 with the incidence of hypoglycemic events in diabetic patients receiving the sulfonylureas glimepiride and gliclazide. Gliclazide 208-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 19891554-9 2009 In a model adjusted for age, BMI, mean daily dose of sulfonylurea, duration of T2DM and renal function, CYP2C9*1/*3 genotype increased the hypoglycemia risk in response to sulfonylurea (odds ratio: 1.687; p = 0.011). Sulfonylurea Compounds 53-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 19891554-9 2009 In a model adjusted for age, BMI, mean daily dose of sulfonylurea, duration of T2DM and renal function, CYP2C9*1/*3 genotype increased the hypoglycemia risk in response to sulfonylurea (odds ratio: 1.687; p = 0.011). Sulfonylurea Compounds 172-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 19891554-11 2009 DISCUSSION & CONCLUSION: The presence of CYP2C9*3 appears to be protective for development of T2DM. Adenosine Monophosphate 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 19891554-12 2009 Furthermore, in T2DM patients, CYP2C9*3 increases the risk of hypoglycemia when they are treated with sulfonylureas, possibly due to impaired metabolism of these drugs. Sulfonylurea Compounds 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 19891554-13 2009 CYP2C9 genotyping might thus be a useful tool for predicting adverse effects caused by sulfonylureas and help clinicians in safer prescribing of oral hypoglycemic agents. Sulfonylurea Compounds 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19484231-0 2009 Severely diminished response to vitamin K-treatment for self-inflicted warfarin intoxication in a patient genotyped as CYP2C9*3*3. Vitamin K 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 19484231-0 2009 Severely diminished response to vitamin K-treatment for self-inflicted warfarin intoxication in a patient genotyped as CYP2C9*3*3. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 19593208-6 2009 A suggestive finding of less pronounced ambulatory BP response to losartan in CYP2C9*1*3 patients with low-normal kidney function was made. Losartan 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 19593208-0 2009 CYP2C9 genotype modifies activity of the renin-angiotensin-aldosterone system in hypertensive men. Aldosterone 59-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 20073138-1 2009 Polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) genes significantly alter the effective warfarin dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 20073138-1 2009 Polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) genes significantly alter the effective warfarin dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 20073138-8 2009 Combinatorial genotyping of CYP2C9 and VKORC1 can allow for individualized dosing of warfarin among patients with gene polymorphisms, potentially reducing the risk of stroke or bleeding. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19578179-12 2009 Thus we confirmed earlier findings that acenocoumarol dosage mainly depends on polymorphisms in the VKORC1 and CYP2C9 genes. Acenocoumarol 40-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 19578179-13 2009 Besides age, gender, body mass index and target INR, one polymorphism within each of the VKORC1, CYP2C9, CYP4F2 and CYP2C18 genes could explain 48.8% of acenocoumarol dosage variation. Acenocoumarol 153-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 19593208-1 2009 BACKGROUND: Two variants of the CYP2C9 gene, CYP2C9*2 and CYP2C9*3, have been indicated to have impaired enzyme function, and thus suspected to reduce the formation of the active metabolite of losartan. Losartan 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 19593208-1 2009 BACKGROUND: Two variants of the CYP2C9 gene, CYP2C9*2 and CYP2C9*3, have been indicated to have impaired enzyme function, and thus suspected to reduce the formation of the active metabolite of losartan. Losartan 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 19593208-1 2009 BACKGROUND: Two variants of the CYP2C9 gene, CYP2C9*2 and CYP2C9*3, have been indicated to have impaired enzyme function, and thus suspected to reduce the formation of the active metabolite of losartan. Losartan 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 19593208-7 2009 At baseline of the GENRES Study, CYP2C9*1*3 patients had significantly lower plasma renin activity and aldosterone levels than CYP2C9*1*1 patients (both P values 0.004). Aldosterone 103-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 19593208-8 2009 In a replication study in patients with treatment-resistant hypertension, men with CYP2C9*3 allele also had lower plasma renin activity (P = 0.03) and aldosterone levels (P = 0.18). Aldosterone 151-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 19593208-10 2009 CONCLUSION: The CYP2C9*3 allele was associated with lower activity of the renin-angiotensin-aldosterone system in hypertensive men, which may reflect a more efficient sodium reabsorption capacity. Aldosterone 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 19593208-10 2009 CONCLUSION: The CYP2C9*3 allele was associated with lower activity of the renin-angiotensin-aldosterone system in hypertensive men, which may reflect a more efficient sodium reabsorption capacity. Sodium 167-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 19752777-1 2009 BACKGROUND: In this prospective cohort study, we have undertaken a comprehensive evaluation of clinical parameters along with variation in 29 genes (including CYP2C9 and VKORC1) to identify factors determining interindividual variability in warfarin response. Warfarin 241-249 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 19604036-0 2009 Effects of the CYP2C9*13 allele on the pharmacokinetics of losartan in healthy male subjects. Losartan 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 19604036-1 2009 The aim of the study was to determine the pharmacokinetics of losartan in relation to the CYP2C9*13 allele. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 19604036-7 2009 Compared with the subjects in the CYP2C9*1/*1 group, individuals carrying the CYP2C9*1/*13 genotype showed significantly a longer t(1/2) of losartan and E3174 and markedly increased the area under the curve (AUC) of losartan. Losartan 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19604036-7 2009 Compared with the subjects in the CYP2C9*1/*1 group, individuals carrying the CYP2C9*1/*13 genotype showed significantly a longer t(1/2) of losartan and E3174 and markedly increased the area under the curve (AUC) of losartan. Losartan 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 19604036-7 2009 Compared with the subjects in the CYP2C9*1/*1 group, individuals carrying the CYP2C9*1/*13 genotype showed significantly a longer t(1/2) of losartan and E3174 and markedly increased the area under the curve (AUC) of losartan. Losartan 216-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19604036-7 2009 Compared with the subjects in the CYP2C9*1/*1 group, individuals carrying the CYP2C9*1/*13 genotype showed significantly a longer t(1/2) of losartan and E3174 and markedly increased the area under the curve (AUC) of losartan. Losartan 216-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 19604036-9 2009 The ratio of AUC(E3174)/AUC(losartan) after losartan administration in the CYP2C9*1/*13 and CYP2C9*1/*3 groups was also statistically different from that in the CYP2C9*1/*1 group. Losartan 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19604036-10 2009 The data indicate that the presence of the CYP2C9*13 allele results in poor metabolism of losartan after a single oral dose. Losartan 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 20031873-1 2009 BACKGROUND: CYP2C9 and VKORC1 genotyping has been advocated as a means of improving the accuracy of warfarin dosing. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19652664-1 2009 Cytochrome P450 2C9 (CYP2C9) enzymes metabolize warfarin and arachidonic acid. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 19652664-1 2009 Cytochrome P450 2C9 (CYP2C9) enzymes metabolize warfarin and arachidonic acid. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 19652664-1 2009 Cytochrome P450 2C9 (CYP2C9) enzymes metabolize warfarin and arachidonic acid. Arachidonic Acid 61-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 19652664-1 2009 Cytochrome P450 2C9 (CYP2C9) enzymes metabolize warfarin and arachidonic acid. Arachidonic Acid 61-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Warfarin 161-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Warfarin 161-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Tolbutamide 173-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Tolbutamide 173-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Phenytoin 186-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Phenytoin 186-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Losartan 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Losartan 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Diclofenac 207-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Diclofenac 207-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Celecoxib 223-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 19925388-1 2009 Human cytochrome P450 2C9 (CYP2C9) accounts for approximately 20% of total hepatic CYP content and metabolizes approximately 15% clinically used drugs including S-warfarin, tolbutamide, phenytoin, losartan, diclofenac, and celecoxib. Celecoxib 223-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19925388-8 2009 Numerous clinical studies have shown that the CYP2C9 polymorphism should be considered in warfarin therapy and practical algorithms how to consider it in therapy are available. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 19925388-11 2009 Polymorphisms in CYP2C9 have the potential to affect the toxicity of CYP2C9 drugs with somewhat lower therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19925388-11 2009 Polymorphisms in CYP2C9 have the potential to affect the toxicity of CYP2C9 drugs with somewhat lower therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19925388-11 2009 Polymorphisms in CYP2C9 have the potential to affect the toxicity of CYP2C9 drugs with somewhat lower therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Phenytoin 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 19925388-11 2009 Polymorphisms in CYP2C9 have the potential to affect the toxicity of CYP2C9 drugs with somewhat lower therapeutic indices such as warfarin, phenytoin, and certain antidiabetic drugs. Phenytoin 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 19541829-6 2009 For diclofenac 4"-hydroxylation, the intrinsic clearance was decreased markedly (by >80%) in CYP2C9.13, CYP2C9.30, and CYP2C9.33 and variably (63-76%) in CYP2C9.3, CYP2C9.26, and CYP2C9.28 due to increased K(m) and/or decreased V(max) values. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19541829-7 2009 For losartan oxidation, CYP2C9.13 and CYP2C9.28 showed 2.5- and 1.8-fold higher K(m) values, respectively, and all variants except for CYP2C9.34 showed >77% lower V(max) and intrinsic clearance values. Losartan 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 19541829-7 2009 For losartan oxidation, CYP2C9.13 and CYP2C9.28 showed 2.5- and 1.8-fold higher K(m) values, respectively, and all variants except for CYP2C9.34 showed >77% lower V(max) and intrinsic clearance values. Losartan 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 19541829-7 2009 For losartan oxidation, CYP2C9.13 and CYP2C9.28 showed 2.5- and 1.8-fold higher K(m) values, respectively, and all variants except for CYP2C9.34 showed >77% lower V(max) and intrinsic clearance values. Losartan 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19541829-8 2009 For glimepiride hydroxylation, the K(m) of CYP2C9.13 was increased 7-fold, and the V(max) values of all variants significantly decreased, resulting in reductions in the intrinsic clearance by >80% in CYP2C9.3, CYP2C9.13, CYP2C9.26, and CYP2C9.33 and by 56 to 75% in CYP2C9.28 and CYP2C9.30. glimepiride 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 19669737-0 2009 CYP2C9 genotype and pharmacodynamic responses to losartan in patients with primary and secondary kidney diseases. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19669737-4 2009 AIM: The study aims were to determine whether CYP2C9 variant alleles (*2 and *3) altered urinary protein excretion, glomerular filtration rate, and blood pressure in Caucasian patients prescribed losartan. Losartan 196-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 19669737-8 2009 CONCLUSION: These preliminary results suggest a possible influence of CYP2C9 genotype on proteinuria and blood pressure in Caucasian CKD patients treated with losartan. Losartan 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 19998810-6 2009 The genetic variants c.430CC and c.1075AA of the CYP2C9 gene were identified, predisposing to rapid warfarin metabolism, as well as the c.-1639GG variant of the VKORC1 gene, associated with low sensitivity to the drug. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 19698014-11 2009 Prasugrel does not appear to interact to any clinically relevant extent with other drugs, including those also metabolized by the hepatic cytochrome P450 isoenzymes CYP3A4, CYP2C9, CYP2C19, and CYP2B6, which are responsible for prasugrel metabolism. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 19761365-4 2009 MATERIALS & METHODS: Seven polymorphic variations in four genes that encode enzymes from phase I (CYP2C9, CYP3A4 and CYP3A5) or phase III (ABCB1) of drug metabolism were analyzed in population samples from Cabinda (n = 107), Mozambique (n = 109) and Sao Tome e Principe (n = 126). Adenosine Monophosphate 11-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 19761366-0 2009 Effects of genetic variation at the CYP2C19/CYP2C9 locus on pharmacokinetics of chlorcycloguanil in adult Gambians. cycloguanil 80-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 19761366-2 2009 MATERIALS & METHODS: Polymorphisms in CYP2C19 and CYP2C9 in 43 adult Gambians treated with chlorproguanil/dapsone for uncomplicated malaria were assessed. Dapsone 110-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 19761366-7 2009 CONCLUSION: CYP2C19*17 determines antimalarial biguanide metabolic profile at the CYP2C19/CYP2C9 locus. Biguanides 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 19694738-1 2009 AIM: To investigate a potential interaction between cranberry juice and diclofenac, a substrate of CYP2C9. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 19706858-9 2009 Thirteen single-nucleotide polymorphisms on chromosome 10q24 within the CYP2C18-CYP2C19-CYP2C9-CYP2C8 cluster were associated with diminished clopidogrel response, with a high degree of statistical significance (P = 1.5 x 10(-13) for rs12777823, additive model). Clopidogrel 142-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19899329-0 2009 CYP2C9 gene analysis of some Iranian hypersensitive patients to warfarin. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19899329-1 2009 For the first time in this study, the pharmacogenetic effects of CYP2C9 polymorphism on warfarin sensitivity in some Iranian patients who are on warfarin treatment were shown. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19899329-1 2009 For the first time in this study, the pharmacogenetic effects of CYP2C9 polymorphism on warfarin sensitivity in some Iranian patients who are on warfarin treatment were shown. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19702548-1 2009 The present study demonstrated that in addition to CYP3A4 and CYP2D6, the metabolism of loratadine is also catalyzed by CYP1A1, CYP2C19, and to a lesser extent by CYP1A2, CYP2B6, CYP2C8, CYP2C9 and CYP3A5. Loratadine 88-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 19448135-6 2009 CYP2D6 coincubation inhibited CYP2C9-mediated (S)-flurbiprofen metabolism in a protein concentration-dependent manner. Flurbiprofen 46-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 19448135-8 2009 Spectral binding studies revealed a 20-fold increase in the K(S) of CYP2C9 toward (S)-flurbiprofen in the presence of CYP2D6. Flurbiprofen 82-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 19454483-1 2009 17alpha-Ethinyl estradiol (EE) was systematically evaluated as a reversible and time-dependent inhibitor of 11 human drug-metabolizing cytochromes P450 (P450s) (CYP1A1, CYP1A2, CYP1B1, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4, and CYP3A5) in vitro. Ethinyl Estradiol 0-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 19453939-1 2009 BACKGROUND: Polymorphisms in the VKORC1 and CYP2C9 genes influence warfarin requirements. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 19297219-0 2009 Influence of CYP2C9 and VKORC1 on warfarin response during initiation of therapy. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 18629445-9 2009 Mean phenprocoumon dosage per week to achieve therapeutic anticoagulation was lower (higher) in patients with than without the VKORC1 polymorphism -1639G > A (3730G > A) or the CYP2C9 polymorphisms. Phenprocoumon 5-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 19663669-1 2009 AIMS: Although the frequencies of pharmacogenetic variants differ among racial groups, most pharmacogenetic algorithms for genotype-guided warfarin dosing only include two CYP2C9 alleles (*2 and *3) and a single VKORC1 allele (g.-1639G>A or g.1173C>T) commonly found among Caucasians. Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 19663669-2 2009 Therefore, this study sought to identify other CYP2C9 and VKORC1 alleles important in warfarin dose variability and to determine their frequencies in different racial and ethnic groups. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 19663669-3 2009 MATERIALS & METHODS: The CYP2C9 and VKORC1 genes were sequenced in selected sensitive (< 21 mg/week) and resistant (> 49 mg/week) individuals with discrepant therapeutic and algorithm-predicted warfarin doses based on prior CYP2C9 and VKORC1 genotyping. Warfarin 204-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19663669-5 2009 RESULTS: Sequencing identified an African-American male with a lower than predicted therapeutic warfarin dose (14.4 mg/week), previously genotyped as CYP2C9*1/*1, who was homozygous for CYP2C9*8 (c.449G>A; p.R150H). Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 19663669-5 2009 RESULTS: Sequencing identified an African-American male with a lower than predicted therapeutic warfarin dose (14.4 mg/week), previously genotyped as CYP2C9*1/*1, who was homozygous for CYP2C9*8 (c.449G>A; p.R150H). Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 19663669-10 2009 Thus, in this racial group, the incorporation of CYP2C9*8 into genotyping panels may improve dose prediction of CYP2C9-metabolized drugs, including warfarin. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 19663669-10 2009 Thus, in this racial group, the incorporation of CYP2C9*8 into genotyping panels may improve dose prediction of CYP2C9-metabolized drugs, including warfarin. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 20140106-3 2009 Achieving safe and effective doses of warfarin therapy is both an urgent and important concern for many clinicians.Recent research has focused on single-nucleotide polymorphisms (SNPs) of genes that encode two proteins: the cytochrome P450 2C9 enzyme and VKORC1 (vitamin K epoxide reductase complex). Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-243 20140106-4 2009 Studies suggest that CYP 2C9 influences warfarin metabolism, whereas VKORC1 plays a role in the pharmacodynamic response in expression of the enzymatic target of warfarin. Warfarin 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-28 20140106-5 2009 Patients who carry CYP 2C9*2 and CYP 2C9*3 alleles tend to require lower warfarin maintenance doses because of their slowed metabolism compared with patients who carry the "wild-type" allele. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-26 20140106-5 2009 Patients who carry CYP 2C9*2 and CYP 2C9*3 alleles tend to require lower warfarin maintenance doses because of their slowed metabolism compared with patients who carry the "wild-type" allele. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-40 19297219-1 2009 BACKGROUND: Although multiple reports have documented the influence of CYP2C9 and VKORC1 variants on warfarin dose, risk of over-anticoagulation and hemorrhage, their influence on anticoagulation maintenance and individual proportion of time spent in target INR range (PPTR) is limited. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 19297219-3 2009 Herein we present the influence of CYP2C9 and VKORC1 C1173T on warfarin response during the first 30 days of therapy. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 19291087-10 2009 We conclude that a NO- and PGI(2)-independent vasodilator mechanism, potentially attributable to EDHF, contributes substantialy to the ACh-induced vasodilatation in human skin microcirculation and that it is probably not a CYP 2C9-derived metabolite. Acetylcholine 135-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-230 19501215-1 2009 BACKGROUND: Etravirine, a non-nucleoside reverse transcriptase inhibitor (NNRTI) active against NNRTI-resistant HIV, is an inducer of CYP3A4 and an inhibitor of CYP2C9/19. etravirine 12-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 19369937-3 2009 In 130 volunteers, CYP2C9 activity was measured in vivo using tolbutamide as a probe drug. Tolbutamide 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 19464879-0 2009 R(+)XK469 inhibits hydroxylation of S-warfarin by CYP2C9. Sulfur 36-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19369937-9 2009 But the difference in activity levels between the CYP2C9*1/*1 and *3/*3 genotypes before the administration of rifampin was sixfold. Rifampin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19799531-5 2009 S-warfarin, predominantly responsible for the anticoagulation effect, is metabolized mostly by the CYP2C9 enzyme. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 19339377-11 2009 Taken together, glucuronidation of cannabinoids depends on upstream processing including enzymes such as CYP2C9 and CYP3A4. Cannabinoids 35-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 19464879-0 2009 R(+)XK469 inhibits hydroxylation of S-warfarin by CYP2C9. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 19464879-1 2009 INTRODUCTION: XK469 is a novel topoisomerase II inhibitor structurally akin to several propionic acid derivatives, such as ibuprofen and diclofenac, which are metabolised by CYP2C9. propionic acid 87-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 19464879-1 2009 INTRODUCTION: XK469 is a novel topoisomerase II inhibitor structurally akin to several propionic acid derivatives, such as ibuprofen and diclofenac, which are metabolised by CYP2C9. Ibuprofen 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 19464879-1 2009 INTRODUCTION: XK469 is a novel topoisomerase II inhibitor structurally akin to several propionic acid derivatives, such as ibuprofen and diclofenac, which are metabolised by CYP2C9. Diclofenac 137-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 19464879-3 2009 The aim of the study is to investigate whether R(+)XK469 interacts with S-warfarin by inhibition of CYP2C9. Warfarin 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 19464879-8 2009 This suggests that coadministration of R(+)XK469 and warfarin results in a clinically significant pharmacokinetic interaction due to CYP2C9 inhibition by R(+)XK469. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 19589229-9 2009 CONCLUSIONS: Fluorophenylpiperazine, methoxyphenylpiperazine, chlorophenylpiperazine, methylbenzylpiperazine and methylenedioxybenzylpiperazine had significant inhibitory effects on CYP2D6, CYP1A2, CYP3A4, CYP2C19 and CYP2C9 isoenzymes but each piperazine had a different inhibitory profile. diethyl 4-methylbenzylphosphonate 86-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 19525490-8 2009 Fluorogenic assays with CYP3A4, CYP2C9, and CYP2D6 on the alginate microarrays exhibited Z" factors ranging from 0.75 to 0.85, sensitive detection of inhibitory compounds, and reactivity comparable to that in solution, thereby demonstrating the reliability and accuracy of the microarray platform. Alginates 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 19589229-9 2009 CONCLUSIONS: Fluorophenylpiperazine, methoxyphenylpiperazine, chlorophenylpiperazine, methylbenzylpiperazine and methylenedioxybenzylpiperazine had significant inhibitory effects on CYP2D6, CYP1A2, CYP3A4, CYP2C19 and CYP2C9 isoenzymes but each piperazine had a different inhibitory profile. fluorophenylpiperazine 13-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 19589229-9 2009 CONCLUSIONS: Fluorophenylpiperazine, methoxyphenylpiperazine, chlorophenylpiperazine, methylbenzylpiperazine and methylenedioxybenzylpiperazine had significant inhibitory effects on CYP2D6, CYP1A2, CYP3A4, CYP2C19 and CYP2C9 isoenzymes but each piperazine had a different inhibitory profile. methoxyphenylpiperazine 37-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 19589229-9 2009 CONCLUSIONS: Fluorophenylpiperazine, methoxyphenylpiperazine, chlorophenylpiperazine, methylbenzylpiperazine and methylenedioxybenzylpiperazine had significant inhibitory effects on CYP2D6, CYP1A2, CYP3A4, CYP2C19 and CYP2C9 isoenzymes but each piperazine had a different inhibitory profile. Piperazine 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 19589229-9 2009 CONCLUSIONS: Fluorophenylpiperazine, methoxyphenylpiperazine, chlorophenylpiperazine, methylbenzylpiperazine and methylenedioxybenzylpiperazine had significant inhibitory effects on CYP2D6, CYP1A2, CYP3A4, CYP2C19 and CYP2C9 isoenzymes but each piperazine had a different inhibitory profile. chlorophenylpiperazine 62-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 19458107-1 2009 BACKGROUND: osartan is metabolized by CYP2C9 and CYP3A4 to an active metabolite, E-3174, which has greater antihypertensive activity than the parent compound. osartan 12-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 19496669-6 2009 RESULTS: A concentration-dependent decrease in luminescence signal was detected for ibuprofen and diclofenac in the assay for CYP 2C9 in human and equine liver microsomes but not in the assay for CYP 3A4 and methadone or xylazine in any of the species. Ibuprofen 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-133 19496669-6 2009 RESULTS: A concentration-dependent decrease in luminescence signal was detected for ibuprofen and diclofenac in the assay for CYP 2C9 in human and equine liver microsomes but not in the assay for CYP 3A4 and methadone or xylazine in any of the species. Diclofenac 98-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-133 19479657-2 2009 The cytochrome P450 (CYP)2C9 and vitamin K epoxide reductase complex 1 (VKORC1) genes have recently been determined to be associated with warfarin dose requirements, with reduced doses of this drug being required in patients with the variant CYP2C9*2, CYP2C9*3, or VKORC1 -1639A allele. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-28 19378397-4 2009 With cisplatin and oxaliplatin, a minor inhibition of CYP2C9 enzyme (75% of control at 400 miromol/l of these complexes) was seen; cisplatin also inhibited slightly the CYP2B6 activity (85% of control). Cisplatin 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 19378397-4 2009 With cisplatin and oxaliplatin, a minor inhibition of CYP2C9 enzyme (75% of control at 400 miromol/l of these complexes) was seen; cisplatin also inhibited slightly the CYP2B6 activity (85% of control). Oxaliplatin 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 19378397-4 2009 With cisplatin and oxaliplatin, a minor inhibition of CYP2C9 enzyme (75% of control at 400 miromol/l of these complexes) was seen; cisplatin also inhibited slightly the CYP2B6 activity (85% of control). miromol 91-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 19378397-9 2009 Interestingly, clinically non-significant inhibition was found with the CYP2C9 and CYP2C8 indicating low probability of interactions with, for example, warfarin. Warfarin 152-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19251823-5 2009 Olomoucine II significantly inhibited the enzymatic activities of CYP1A2, CYP2C9, and (to a lesser degree) CYP3A4. olomoucine II 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 19251823-6 2009 The results indicate that use of olomoucine II as a drug could affect the activities of CYP3A4, CYP1A2, and CYP2C9 in vivo. olomoucine II 33-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 19221727-4 2009 RESULTS: The area under the plasma concentration-time curve (AUC) of losartan increased significantly following a 14-day silymarin treatment in subjects with the CYP2C9*1/*1 genotype, but not in those with the CYP2C9*1/*3 genotype. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19221727-4 2009 RESULTS: The area under the plasma concentration-time curve (AUC) of losartan increased significantly following a 14-day silymarin treatment in subjects with the CYP2C9*1/*1 genotype, but not in those with the CYP2C9*1/*3 genotype. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 210-216 19221727-4 2009 RESULTS: The area under the plasma concentration-time curve (AUC) of losartan increased significantly following a 14-day silymarin treatment in subjects with the CYP2C9*1/*1 genotype, but not in those with the CYP2C9*1/*3 genotype. Silymarin 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19221727-5 2009 The AUC of E-3174 decreased significantly with a silymarin pretreatment in both CYP2C9*1/*1 and the CYP2C9*1/*3 subjects. losartan carboxylic acid 11-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 19221727-5 2009 The AUC of E-3174 decreased significantly with a silymarin pretreatment in both CYP2C9*1/*1 and the CYP2C9*1/*3 subjects. losartan carboxylic acid 11-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 19221727-5 2009 The AUC of E-3174 decreased significantly with a silymarin pretreatment in both CYP2C9*1/*1 and the CYP2C9*1/*3 subjects. Silymarin 49-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 19221727-5 2009 The AUC of E-3174 decreased significantly with a silymarin pretreatment in both CYP2C9*1/*1 and the CYP2C9*1/*3 subjects. Silymarin 49-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 19221727-6 2009 The metabolic ratio of losartan (ratio of AUC(0-infinity) of E-3174 to AUC(0-infinity) of losartan) decreased significantly after a 14-day treatment with silymarin in individuals with the CYP2C9*1/*1 genotype (p < 0.05), but not in those with the CYP2C9*1/*3 genotype (p = 0.065). Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 19221727-6 2009 The metabolic ratio of losartan (ratio of AUC(0-infinity) of E-3174 to AUC(0-infinity) of losartan) decreased significantly after a 14-day treatment with silymarin in individuals with the CYP2C9*1/*1 genotype (p < 0.05), but not in those with the CYP2C9*1/*3 genotype (p = 0.065). Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 250-256 19221727-6 2009 The metabolic ratio of losartan (ratio of AUC(0-infinity) of E-3174 to AUC(0-infinity) of losartan) decreased significantly after a 14-day treatment with silymarin in individuals with the CYP2C9*1/*1 genotype (p < 0.05), but not in those with the CYP2C9*1/*3 genotype (p = 0.065). Silymarin 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 19221727-6 2009 The metabolic ratio of losartan (ratio of AUC(0-infinity) of E-3174 to AUC(0-infinity) of losartan) decreased significantly after a 14-day treatment with silymarin in individuals with the CYP2C9*1/*1 genotype (p < 0.05), but not in those with the CYP2C9*1/*3 genotype (p = 0.065). Silymarin 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 250-256 19221727-7 2009 CONCLUSION: Silymarin inhibits the metabolism of losartan to E-3174, with the magnitude of the interaction differing in individuals with different CYP2C9 genotypes. Silymarin 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 19479657-2 2009 The cytochrome P450 (CYP)2C9 and vitamin K epoxide reductase complex 1 (VKORC1) genes have recently been determined to be associated with warfarin dose requirements, with reduced doses of this drug being required in patients with the variant CYP2C9*2, CYP2C9*3, or VKORC1 -1639A allele. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 242-248 19479657-2 2009 The cytochrome P450 (CYP)2C9 and vitamin K epoxide reductase complex 1 (VKORC1) genes have recently been determined to be associated with warfarin dose requirements, with reduced doses of this drug being required in patients with the variant CYP2C9*2, CYP2C9*3, or VKORC1 -1639A allele. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 252-258 19479657-4 2009 Racial differences in the association between genotype and a patient"s response to warfarin treatment may be caused by racial differences in the frequencies of the variant CYP2C9*2, CYP2C9*3, and VKORC1 -1639A alleles or by the influence of non-genetic factors. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 19233181-0 2009 Cytochrome P450 2C9 variants influence response to celecoxib for prevention of colorectal adenoma. Celecoxib 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 19479657-4 2009 Racial differences in the association between genotype and a patient"s response to warfarin treatment may be caused by racial differences in the frequencies of the variant CYP2C9*2, CYP2C9*3, and VKORC1 -1639A alleles or by the influence of non-genetic factors. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 19233181-1 2009 BACKGROUND & AIMS: Variants in the cytochrome P450 2C9 (CYP2C9) gene are associated with impaired metabolism of celecoxib. Adenosine Monophosphate 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 19258521-2 2009 Although it is well established that CYP2C9 is the major cytochrome P450 enzyme controlling metabolic elimination of phenytoin through its oxidative conversion to (S)-5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH), nothing is known about the amino acid binding determinants within the CYP2C9 active site that promote metabolism and maintain the tight stereocontrol of hydroxy metabolite formation. p-hpph 206-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19233181-1 2009 BACKGROUND & AIMS: Variants in the cytochrome P450 2C9 (CYP2C9) gene are associated with impaired metabolism of celecoxib. Adenosine Monophosphate 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 19233181-1 2009 BACKGROUND & AIMS: Variants in the cytochrome P450 2C9 (CYP2C9) gene are associated with impaired metabolism of celecoxib. Celecoxib 116-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 19233181-1 2009 BACKGROUND & AIMS: Variants in the cytochrome P450 2C9 (CYP2C9) gene are associated with impaired metabolism of celecoxib. Celecoxib 116-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 19233181-10 2009 CONCLUSIONS: The greater efficacy of high-dose celecoxib, compared with the low-dose, in preventing colorectal adenoma appears confined to individuals with slow metabolizer (CYP2C9*3) genotypes. Celecoxib 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. aceclofenac 169-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. aceclofenac 169-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Celecoxib 182-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Celecoxib 182-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Diclofenac 193-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Diclofenac 193-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Ibuprofen 205-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Ibuprofen 205-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Indomethacin 216-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Indomethacin 216-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. lornoxicam 231-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. lornoxicam 231-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Meloxicam 243-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Meloxicam 243-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Naproxen 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Naproxen 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Piroxicam 264-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. Piroxicam 264-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. tenoxicam 275-284 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. tenoxicam 275-284 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. valdecoxib 289-299 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19422321-1 2009 Polymorphisms in CYP2C8 and CYP2C9 are common in all the human populations and many CYP2C8 and CYP2C9 gene variations cause decreased enzyme activity towards the NSAIDs aceclofenac, celecoxib, diclofenac, ibuprofen, indomethazine, lornoxicam, meloxicam, naproxen, piroxicam, tenoxicam and valdecoxib. valdecoxib 289-299 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 19258521-0 2009 CYP2C9 amino acid residues influencing phenytoin turnover and metabolite regio- and stereochemistry. Phenytoin 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19258521-2 2009 Although it is well established that CYP2C9 is the major cytochrome P450 enzyme controlling metabolic elimination of phenytoin through its oxidative conversion to (S)-5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH), nothing is known about the amino acid binding determinants within the CYP2C9 active site that promote metabolism and maintain the tight stereocontrol of hydroxy metabolite formation. Phenytoin 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19258521-2 2009 Although it is well established that CYP2C9 is the major cytochrome P450 enzyme controlling metabolic elimination of phenytoin through its oxidative conversion to (S)-5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH), nothing is known about the amino acid binding determinants within the CYP2C9 active site that promote metabolism and maintain the tight stereocontrol of hydroxy metabolite formation. Phenytoin 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 285-291 19258521-2 2009 Although it is well established that CYP2C9 is the major cytochrome P450 enzyme controlling metabolic elimination of phenytoin through its oxidative conversion to (S)-5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH), nothing is known about the amino acid binding determinants within the CYP2C9 active site that promote metabolism and maintain the tight stereocontrol of hydroxy metabolite formation. hydroxyphenytoin 163-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19258521-2 2009 Although it is well established that CYP2C9 is the major cytochrome P450 enzyme controlling metabolic elimination of phenytoin through its oxidative conversion to (S)-5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH), nothing is known about the amino acid binding determinants within the CYP2C9 active site that promote metabolism and maintain the tight stereocontrol of hydroxy metabolite formation. p-hpph 206-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 285-291 19258521-5 2009 This pattern of effects differs substantially from that found previously for (S)-warfarin and (S)-flurbiprofen metabolism, suggesting that these three ligands bind within discrete locations in the CYP2C9 active site. Warfarin 77-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 19258521-5 2009 This pattern of effects differs substantially from that found previously for (S)-warfarin and (S)-flurbiprofen metabolism, suggesting that these three ligands bind within discrete locations in the CYP2C9 active site. Flurbiprofen 94-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 19662888-2 2009 The application of pharmacogenetics in testing individual polymorphisms of two genes CYP 2C9 (pharmacokinetics of warfarin) and VKORC1 (sensitivity on warfarin) is promising tactics leading to a safe anticoagulation. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-92 19501186-6 2009 Among these transformants, benzo(a)pyrene-induced or cyclophosphamide-produced micronucleus (MN) frequency was markedly increased in transformants expressing CYP1A2 or CYP2C9, respectively. Benzo(a)pyrene 27-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 19501186-6 2009 Among these transformants, benzo(a)pyrene-induced or cyclophosphamide-produced micronucleus (MN) frequency was markedly increased in transformants expressing CYP1A2 or CYP2C9, respectively. Cyclophosphamide 53-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 19480553-0 2009 Pharmacokinetic studies of enantiomers of ibuprofen and its chiral metabolites in humans with different variants of genes coding CYP2C8 and CYP2C9 isoenzymes. Ibuprofen 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 19480553-5 2009 Impaired metabolism of ibuprofen enantiomers was associated with the presence of CYP2C8*3, CYP2C9*2 and CYP2C9*3 alleles. Ibuprofen 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19480553-5 2009 Impaired metabolism of ibuprofen enantiomers was associated with the presence of CYP2C8*3, CYP2C9*2 and CYP2C9*3 alleles. Ibuprofen 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 19297519-1 2009 Genetic polymorphisms in VKORC1 and CYP2C9, genes controlling vitamin K(1) (VK1) epoxide reduction and (S)-warfarin metabolism, respectively, are major contributors to interindividual variability in warfarin dose. Vitamin K 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 19297519-1 2009 Genetic polymorphisms in VKORC1 and CYP2C9, genes controlling vitamin K(1) (VK1) epoxide reduction and (S)-warfarin metabolism, respectively, are major contributors to interindividual variability in warfarin dose. Epoxy Compounds 81-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 19297519-1 2009 Genetic polymorphisms in VKORC1 and CYP2C9, genes controlling vitamin K(1) (VK1) epoxide reduction and (S)-warfarin metabolism, respectively, are major contributors to interindividual variability in warfarin dose. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 19297519-1 2009 Genetic polymorphisms in VKORC1 and CYP2C9, genes controlling vitamin K(1) (VK1) epoxide reduction and (S)-warfarin metabolism, respectively, are major contributors to interindividual variability in warfarin dose. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 19662888-2 2009 The application of pharmacogenetics in testing individual polymorphisms of two genes CYP 2C9 (pharmacokinetics of warfarin) and VKORC1 (sensitivity on warfarin) is promising tactics leading to a safe anticoagulation. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-92 19255811-1 2009 BACKGROUND: Genetic variants of the warfarin sensitivity gene CYP2C9 have been associated with increased bleeding risk during warfarin initiation. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19245785-6 2009 CYP2C92 catalyzed diclofenac metabolism was 20-fold slower than the human counterpart, CYP2C9. Diclofenac 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19245785-7 2009 CYP2C92 demonstrated comparable tolbutamide and (S)-warfarin hydroxylase activity compared to CYP2C9, upon addition of b(5) to the reactions. Tolbutamide 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19196847-3 2009 Relative activity factors were measured by determining the intrinsic clearance (CL(int)) of probe substrates (bufuralol-CYP2D6, diclofenac-CYP2C9, midazolam-CYP3A4, and phenacetin-CYP1A2) in recombinant P450s and 16 HLM donors. Diclofenac 128-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 19255811-9 2009 RESULTS: In the base case, testing and treating patients with CYP2C9*2 and/or CYP2C9*3 with aspirin rather than warfarin was best (8.97 QALYs). Aspirin 92-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 19255811-1 2009 BACKGROUND: Genetic variants of the warfarin sensitivity gene CYP2C9 have been associated with increased bleeding risk during warfarin initiation. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19255811-9 2009 RESULTS: In the base case, testing and treating patients with CYP2C9*2 and/or CYP2C9*3 with aspirin rather than warfarin was best (8.97 QALYs). Aspirin 92-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19495518-0 2009 CYP2C9 polymorphism: prevalence in healthy and warfarin-treated Malay and Chinese in Malaysia. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19442216-3 2009 In order to understand the metabolic mechanism of various drugs, two crystal structures of CYP2C9 have been studied, and their structural differences and structure-activity relationships with the drugs of Fluoxetine, Ibuprofen, Naproxen, Suprofen, and Mefenamic acid were investigated. Fluoxetine 205-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19442216-3 2009 In order to understand the metabolic mechanism of various drugs, two crystal structures of CYP2C9 have been studied, and their structural differences and structure-activity relationships with the drugs of Fluoxetine, Ibuprofen, Naproxen, Suprofen, and Mefenamic acid were investigated. Ibuprofen 217-226 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19442216-3 2009 In order to understand the metabolic mechanism of various drugs, two crystal structures of CYP2C9 have been studied, and their structural differences and structure-activity relationships with the drugs of Fluoxetine, Ibuprofen, Naproxen, Suprofen, and Mefenamic acid were investigated. Naproxen 228-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19442216-3 2009 In order to understand the metabolic mechanism of various drugs, two crystal structures of CYP2C9 have been studied, and their structural differences and structure-activity relationships with the drugs of Fluoxetine, Ibuprofen, Naproxen, Suprofen, and Mefenamic acid were investigated. Suprofen 238-246 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19442216-3 2009 In order to understand the metabolic mechanism of various drugs, two crystal structures of CYP2C9 have been studied, and their structural differences and structure-activity relationships with the drugs of Fluoxetine, Ibuprofen, Naproxen, Suprofen, and Mefenamic acid were investigated. Mefenamic Acid 252-266 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 19324988-1 2009 Responses to warfarin (Coumadin) anticoagulation therapy are affected by genetic variability in both the CYP2C9 and VKORC1 genes. Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 19324988-1 2009 Responses to warfarin (Coumadin) anticoagulation therapy are affected by genetic variability in both the CYP2C9 and VKORC1 genes. Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 19324988-3 2009 We compared four platforms for determining the relevant single nucleotide polymorphisms (SNPs) in both CYP2C9 and VKORC1 that are associated with warfarin sensitivity (Third Wave Invader Plus, ParagonDx/Cepheid Smart Cycler, Idaho Technology LightCycler, and AutoGenomics Infiniti). Warfarin 146-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 19495518-2 2009 CYP2C9 has been reported to be the enzyme responsible for the metabolism of many drugs, including warfarin and other drugs with a narrow therapeutic index. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19495518-4 2009 We aimed to evaluate the prevalence of the types and frequencies of common CYP2C9 alleles (*1, *2, *3 and *4) among the healthy unrelated individuals and diseased patients prescribed with warfarin. Warfarin 188-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19495518-12 2009 Among the warfarin-treated group, only CYP2C9*1 and *3 were detected. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 19495518-14 2009 CONCLUSION: Based on the above-observed genotypes, the prevalence of CYP2C9*2 and *3 was low in healthy and warfarin-treated Malays and Chinese in Malaysia. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 19225451-1 2009 The objective of this study was to investigate the influence of genotypes associated with reduced activity of vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) on anticoagulation with acenocoumarol during the first 6 weeks of treatment. Acenocoumarol 222-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-188 19022234-6 2009 By co-treatment with specific inhibitors for cytochrome P450 (CYP) 2C and CYP3A - the major phase I enzymes involved in liver xenobiotic metabolism - we could confirm the prominent role of CYP3A in the bioactivation of troglitazone as well as the role of CYP3A and CYP2C in the activation of diclofenac. Troglitazone 219-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-69 19022234-6 2009 By co-treatment with specific inhibitors for cytochrome P450 (CYP) 2C and CYP3A - the major phase I enzymes involved in liver xenobiotic metabolism - we could confirm the prominent role of CYP3A in the bioactivation of troglitazone as well as the role of CYP3A and CYP2C in the activation of diclofenac. Diclofenac 292-302 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-69 19233910-0 2009 A multiplex assay for detecting genetic variations in CYP2C9, VKORC1, and GGCX involved in warfarin metabolism. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 19233910-2 2009 Genetic variations in the cytochrome P450, family 2, subfamily C, polypeptide 9 (CYP2C9), vitamin K epoxide reductase complex, subunit 1 (VKORC1), and gamma-glutamyl carboxylase (GGCX) genes have been shown to contribute to impaired metabolism of warfarin. Warfarin 247-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-79 19074728-0 2009 Relative contribution of CYP2C9 and VKORC1 genotypes and early INR response to the prediction of warfarin sensitivity during initiation of therapy. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19074728-1 2009 Genetic variants in CYP2C9 and VKORC1 strongly affect steady-state warfarin dose. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 19074728-4 2009 In 214 patients starting warfarin with INR-guided dose adjustments, we determined whether CYP2C9 and VKORC1 genotypes were associated with early measures of warfarin sensitivity (time to INR >/= lower limit of therapeutic range; time to INR > 4; and first stable warfarin dose) after adjusting for early (days 4-6) and week 1 (days 7-9) INR values. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 19074728-4 2009 In 214 patients starting warfarin with INR-guided dose adjustments, we determined whether CYP2C9 and VKORC1 genotypes were associated with early measures of warfarin sensitivity (time to INR >/= lower limit of therapeutic range; time to INR > 4; and first stable warfarin dose) after adjusting for early (days 4-6) and week 1 (days 7-9) INR values. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 19074728-9 2009 Thus, much of the information provided by CYP2C9 and VKORC1 genotypes during warfarin initiation is captured by the early INR response. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 18950609-12 2009 Results with the isoform-selective inhibitors suggest that the isoforms inhibited by ketoconazole (mainly CYP3A4) and diethyldithiocarbamate (mainly CYP2A6, and CYP2E1), but not the isoforms inhibited by furafylline (mainly CYP1A2), sulfaphenazole (mainly CYP2C9) and quinidine (mainly CYP2D6) are involved in the metabolic activation of AFB1. Ketoconazole 85-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 256-262 18950609-12 2009 Results with the isoform-selective inhibitors suggest that the isoforms inhibited by ketoconazole (mainly CYP3A4) and diethyldithiocarbamate (mainly CYP2A6, and CYP2E1), but not the isoforms inhibited by furafylline (mainly CYP1A2), sulfaphenazole (mainly CYP2C9) and quinidine (mainly CYP2D6) are involved in the metabolic activation of AFB1. Ditiocarb 118-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 256-262 19181737-0 2009 Rapid single-nucleotide polymorphism detection of cytochrome P450 (CYP2C9) and vitamin K epoxide reductase (VKORC1) genes for the warfarin dose adjustment by the SMart-amplification process version 2. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 19181737-1 2009 BACKGROUND: Polymorphisms of the CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) gene (CYP2C9*2, CYP2C9*3) and the VKORC1 (vitamin K epoxide reductase complex, subunit 1) gene (-1639G>A) greatly impact the maintenance dose for the drug warfarin. Warfarin 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 19181737-1 2009 BACKGROUND: Polymorphisms of the CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) gene (CYP2C9*2, CYP2C9*3) and the VKORC1 (vitamin K epoxide reductase complex, subunit 1) gene (-1639G>A) greatly impact the maintenance dose for the drug warfarin. Warfarin 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-94 19181737-1 2009 BACKGROUND: Polymorphisms of the CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) gene (CYP2C9*2, CYP2C9*3) and the VKORC1 (vitamin K epoxide reductase complex, subunit 1) gene (-1639G>A) greatly impact the maintenance dose for the drug warfarin. Warfarin 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 19181737-1 2009 BACKGROUND: Polymorphisms of the CYP2C9 (cytochrome P450, family 2, subfamily C, polypeptide 9) gene (CYP2C9*2, CYP2C9*3) and the VKORC1 (vitamin K epoxide reductase complex, subunit 1) gene (-1639G>A) greatly impact the maintenance dose for the drug warfarin. Warfarin 254-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 19225451-1 2009 The objective of this study was to investigate the influence of genotypes associated with reduced activity of vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) on anticoagulation with acenocoumarol during the first 6 weeks of treatment. Acenocoumarol 222-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 19225451-6 2009 Our conclusion was that an initial standard dosing regimen with acenocoumarol increases the risk of severe overanticoagulation in patients with variant alleles of the VKORC1 and CYP2C9 genes. Acenocoumarol 64-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 19031075-0 2009 Influence of CYP2C9 genotype on warfarin dose requirements--a systematic review and meta-analysis. Warfarin 32-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19031075-1 2009 PURPOSE: To quantify the influence of common cytochrome P450 2C9 (CYP2C9) polymorphisms on warfarin dose requirements. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-64 19031075-1 2009 PURPOSE: To quantify the influence of common cytochrome P450 2C9 (CYP2C9) polymorphisms on warfarin dose requirements. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 19031075-2 2009 METHODS: A systematic review and a meta-analysis, calculating the warfarin dose reduction associated with the five most common variant CYP2C9 genotypes. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 19031075-6 2009 CONCLUSIONS: Previous studies have rarely been powered to determine the quantitative influence of specific CYP2C9 genotypes on warfarin dose requirements. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 19442086-6 2009 CYP2B6, CYP3A4 and CYP2C9 play a relevant role in the metabolism of ketamine. Ketamine 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 19171677-2 2009 To address whether the need for redox transfer proteins and the NADPH cofactor protein could be obviated, CYP2C9 was bonded to a gold electrode through an 11-mercaptoundecanoic acid and octanethiol self-assembled monolayer (SAM) through which a current could be applied. 11-mercaptoundecanoic acid 155-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 19171677-2 2009 To address whether the need for redox transfer proteins and the NADPH cofactor protein could be obviated, CYP2C9 was bonded to a gold electrode through an 11-mercaptoundecanoic acid and octanethiol self-assembled monolayer (SAM) through which a current could be applied. n-octanethiol 186-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 19171677-3 2009 Cyclic voltammetry demonstrated direct electrochemistry of the CYP2C9 enzyme bonded to the electrode and fast electron transfer between the heme iron and the gold electrode. Heme 140-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19171677-3 2009 Cyclic voltammetry demonstrated direct electrochemistry of the CYP2C9 enzyme bonded to the electrode and fast electron transfer between the heme iron and the gold electrode. Iron 145-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19171677-4 2009 To determine whether this system could metabolize warfarin analogous to microsomal or expressed enzyme systems containing CYP2C9, warfarin was incubated with the CYP2C9-SAM-gold electrode and a controlled potential was applied. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 19171677-5 2009 The expected 7-hydroxywarfarin metabolite was observed, analogous to expressed CYP2C9 systems, wherein this is the predominant metabolite. 7-hydroxywarfarin 13-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19171677-7 2009 In summary, the CYP2C9-SAM-gold electrode system was able to carry out the metabolism of warfarin only after application of an electrical potential, but in the absence of either CPR or NADPH. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 19083245-0 2009 A potentially deleterious new CYP2C9 polymorphism identified in an African American patient with major hemorrhage on warfarin therapy. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 19005461-0 2009 Interindividual variation in the pharmacokinetics of Delta9-tetrahydrocannabinol as related to genetic polymorphisms in CYP2C9. Dronabinol 53-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 19005461-1 2009 The impact of the CYP2C9 polymorphism on the pharmacokinetics of orally administered 9-tetrahydrocannabinol (THC) was studied in 43 healthy volunteers. Dronabinol 85-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 19005461-3 2009 However, the median area under the curve of THC was threefold higher and that of 11-nor-9-carboxy-9-tetrahydrocannabinol was 70% lower in CYP2C9*3/*3 homozygotes than in CYP2C9*1/*1 homozygotes. Dronabinol 44-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 19005461-3 2009 However, the median area under the curve of THC was threefold higher and that of 11-nor-9-carboxy-9-tetrahydrocannabinol was 70% lower in CYP2C9*3/*3 homozygotes than in CYP2C9*1/*1 homozygotes. 11-nor-9-carboxy-9-tetrahydrocannabinol 81-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 19005461-3 2009 However, the median area under the curve of THC was threefold higher and that of 11-nor-9-carboxy-9-tetrahydrocannabinol was 70% lower in CYP2C9*3/*3 homozygotes than in CYP2C9*1/*1 homozygotes. 11-nor-9-carboxy-9-tetrahydrocannabinol 81-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 19005461-4 2009 CYP2C9*3 carriers also showed a trend toward increased sedation following administration of THC. Dronabinol 92-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19005461-5 2009 Therefore, the CYP2C9*3 variant may influence both the therapeutic and adverse effects of THC. Dronabinol 90-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 19601875-3 2009 Glutathione and cyanide did not inhibit the enzymes at concentrations up to 10 mM; however methoxylamine did show inhibition, with IC(50) values of 0.53 mM for CYP1A2, 4.12 mM for CYP2C9, 2.04 mM for CYP2C19, 9.72 mM for CYP2D6, and 1.26 and >10 mM for CYP3A4/5 (for testosterone and midazolam, respectively, as substrates). methoxyamine 91-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 19289643-3 2009 We aimed to assess the effect of sulfaphenazole, a specific CYP 2C9 inhibitor, on tPA release in normotensive subjects and patients with essential hypertension. Sulfaphenazole 33-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-67 19289643-11 2009 CONCLUSIONS: Sulfaphenazole inhibits bradykinin-induced tPA release, which suggests a modulatory role of CYP 2C9-derived endothelium-derived hyperpolarizing factors in tPA release in humans. Sulfaphenazole 13-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-112 19252308-0 2009 Effect of ethanol on S-warfarin and diclofenac metabolism by recombinant human CYP2C9.1. Ethanol 10-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19252308-0 2009 Effect of ethanol on S-warfarin and diclofenac metabolism by recombinant human CYP2C9.1. Warfarin 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19252308-0 2009 Effect of ethanol on S-warfarin and diclofenac metabolism by recombinant human CYP2C9.1. Diclofenac 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 19252308-1 2009 The effect of ethanol on the metabolism of S-warfarin and diclofenac by recombinant cytochrome P450 2C9.1 microsomes (CYP2C9.1) was studied. Ethanol 14-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 19252308-1 2009 The effect of ethanol on the metabolism of S-warfarin and diclofenac by recombinant cytochrome P450 2C9.1 microsomes (CYP2C9.1) was studied. Warfarin 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 19252308-1 2009 The effect of ethanol on the metabolism of S-warfarin and diclofenac by recombinant cytochrome P450 2C9.1 microsomes (CYP2C9.1) was studied. Warfarin 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 19252308-1 2009 The effect of ethanol on the metabolism of S-warfarin and diclofenac by recombinant cytochrome P450 2C9.1 microsomes (CYP2C9.1) was studied. Diclofenac 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 19252308-1 2009 The effect of ethanol on the metabolism of S-warfarin and diclofenac by recombinant cytochrome P450 2C9.1 microsomes (CYP2C9.1) was studied. Diclofenac 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 19252308-5 2009 Ethanol at a concentration of 3.0 vol% (510 mM) increased the K(m) value of diclofenac metabolism without changes in the V(max), which indicates that diclofenac 4"-hydroxylation by CYP2C9.1 was competitively inhibited by ethanol. Ethanol 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 19252308-5 2009 Ethanol at a concentration of 3.0 vol% (510 mM) increased the K(m) value of diclofenac metabolism without changes in the V(max), which indicates that diclofenac 4"-hydroxylation by CYP2C9.1 was competitively inhibited by ethanol. Diclofenac 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 19252308-5 2009 Ethanol at a concentration of 3.0 vol% (510 mM) increased the K(m) value of diclofenac metabolism without changes in the V(max), which indicates that diclofenac 4"-hydroxylation by CYP2C9.1 was competitively inhibited by ethanol. Diclofenac 150-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 19252308-5 2009 Ethanol at a concentration of 3.0 vol% (510 mM) increased the K(m) value of diclofenac metabolism without changes in the V(max), which indicates that diclofenac 4"-hydroxylation by CYP2C9.1 was competitively inhibited by ethanol. Ethanol 221-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 19252308-6 2009 S-Warfarin metabolism by CYP2C9.1 was more sensitive to ethanol than diclofenac metabolism. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19252308-6 2009 S-Warfarin metabolism by CYP2C9.1 was more sensitive to ethanol than diclofenac metabolism. Ethanol 56-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19252308-7 2009 These results suggest that ethanol inhibits the metabolism by CYP2C9.1 in a substrate-dependent manner. Ethanol 27-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19074529-0 2009 Mechanism of CYP2C9 inhibition by flavones and flavonols. Flavones 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19074529-0 2009 Mechanism of CYP2C9 inhibition by flavones and flavonols. Flavonols 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19074529-2 2009 With the exception of flavone, all flavonoids were shown to inhibit CYP2C9-mediated diclofenac 4"-hydroxylation in the CYP2C9 RECO system, with K(i) value <or= 2.2 microM. Flavonoids 35-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 19074529-2 2009 With the exception of flavone, all flavonoids were shown to inhibit CYP2C9-mediated diclofenac 4"-hydroxylation in the CYP2C9 RECO system, with K(i) value <or= 2.2 microM. Flavonoids 35-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 19074529-2 2009 With the exception of flavone, all flavonoids were shown to inhibit CYP2C9-mediated diclofenac 4"-hydroxylation in the CYP2C9 RECO system, with K(i) value <or= 2.2 microM. Diclofenac 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 19074529-2 2009 With the exception of flavone, all flavonoids were shown to inhibit CYP2C9-mediated diclofenac 4"-hydroxylation in the CYP2C9 RECO system, with K(i) value <or= 2.2 microM. Diclofenac 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 19074529-3 2009 In terms of the mechanism of inhibition, 6-hydroxyflavone was found to be a noncompetitive inhibitor of CYP2C9, whereas the other flavonoids were competitive inhibitors. 6-hydroxyflavone 41-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 19074529-4 2009 Computer docking simulation and constructed mutants substituted at residue 100 of CYP2C9.1 indicate that the noncompetitive binding site of 6-hydroxyflavone lies beside Phe100, similar to the reported allosteric binding site of warfarin. 6-hydroxyflavone 140-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 19074529-4 2009 Computer docking simulation and constructed mutants substituted at residue 100 of CYP2C9.1 indicate that the noncompetitive binding site of 6-hydroxyflavone lies beside Phe100, similar to the reported allosteric binding site of warfarin. Warfarin 228-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 19074529-5 2009 The other flavonoids exert competitive inhibition through interaction with the substrate binding site of CYP2C9 accessed by flurbiprofen. Flavonoids 10-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 19074529-5 2009 The other flavonoids exert competitive inhibition through interaction with the substrate binding site of CYP2C9 accessed by flurbiprofen. Flurbiprofen 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 19074529-6 2009 These results suggest flavonoids can participate in interactions with drugs that act as substrates for CYP2C9 and provide a possible molecular basis for understanding cooperativity in human P450-mediated drug-drug interactions. Flavonoids 22-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 19442086-8 2009 Propofol is metabolized mainly by glucuronidation by uridine diphosphate-glucuronosyltransferases (UGTs) and by hydroxylation by CYP2B6 and CYP2C enzymes. Propofol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-145 19022226-6 2009 At 10 microM PCEEA, CBP reduced metabolite formation by 61%, while inhibition of CYP3A4 by ketoconazole and inhibition of CYP2C9 by sulfaphenazole showed no inhibitory effect. Sulfaphenazole 132-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 19300499-0 2009 A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 19300499-5 2009 We initially tested each of 325,997 GWAS SNPs for association with warfarin dose by univariate regression and found the strongest statistical signals (p<10(-78)) at SNPs clustering near VKORC1 and the second lowest p-values (p<10(-31)) emanating from CYP2C9. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 257-263 19277427-0 2009 Pharmacogenetics of acenocoumarol: CYP2C9 *2 and VKORC1 c.-1639G>A, 497C>G, 1173C>T, and 3730G>A variants influence drug dose in anticoagulated patients. Acenocoumarol 20-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 19337788-0 2009 Frequency of CYP3A4, CYP3A5, CYP2C9, and CYP2C19 variant alleles in patients receiving clopidogrel that experience repeat acute coronary syndrome. Clopidogrel 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19337788-2 2009 This research evaluated the frequency of variant alleles in the genes coding for CYP3A4, CYP3A5, CYP2C9, and CYP2C19 enzymes in patients on clopidogrel therapy and experiencing repeat acute coronary syndrome (ACS) compared to a control group with a matching ethnic composition. Clopidogrel 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 19177029-0 2009 Validation of VKORC1 and CYP2C9 genotypes on interindividual warfarin maintenance dose: a prospective study in Chinese patients. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 19029318-5 2009 In pilot experiments, voriconazole showed inhibition of CYP2B6, CYP2C9, CYP2C19, and CYP3A (half-maximal [50%] inhibitory concentrations, <6 microM); its effect on CYP1A2, CYP2A6, CYP2C8, and CYP2D6 was marginal (<25% inhibition at 100 microM voriconazole). Voriconazole 22-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 19029318-6 2009 Further detailed experiments with HLMs showed that voriconazole is a potent competitive inhibitor of CYP2B6 (K(i) < 0.5), CYP2C9 (K(i) = 2.79 microM), and CYP2C19 (K(i) = 5.1 microM). Voriconazole 51-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 19029318-8 2009 Prediction of the in vivo interaction of voriconazole from these in vitro data suggests that voriconazole would substantially increase the exposure of drugs metabolized by CYP2B6, CYP2C9, CYP2C19, and CYP3A. Voriconazole 41-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 19029318-8 2009 Prediction of the in vivo interaction of voriconazole from these in vitro data suggests that voriconazole would substantially increase the exposure of drugs metabolized by CYP2B6, CYP2C9, CYP2C19, and CYP3A. Voriconazole 93-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 19280158-0 2009 Effect of CYP2C9*3 allele on the pharmacokinetics of naproxen in Korean subjects. Naproxen 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 19095442-3 2009 These studies culminated in the identification of anthranilimides 16 and 22 which displayed potent in vitro inhibition of GPa in addition to reduced inhibition of CYP2C9 and excellent pharmacokinetic properties. anthranilimides 50-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 19280158-1 2009 The genetically polymorphic CYP2C9 metabolizes many non-steroidal anti-inflammatory agents, including naproxen. Naproxen 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19280158-2 2009 This study examined the effects of a CYP2C9 genetic polymorphism on the pharmacokinetics of naproxen in Korean subjects. Naproxen 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19280158-6 2009 The mean plasma concentration-time profiles of naproxen in the CYP2C9*1/*3 and CYP2C9*1/*1 individuals were similar. Naproxen 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 19196837-8 2009 The mechanism of interaction between antiretroviral agents and warfarin appears to be mediated through alteration in CYP2C9 metabolism. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 18971317-9 2009 Rosuvastatin was found to increase human constitutive androstane receptor (hCAR)-mediated transcription of CYP3A4, CYP2C9, and CYP2B6 genes, predicting the consequent potential for drug interactions with several coadministered drugs. Rosuvastatin Calcium 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 18971317-10 2009 Activation of hCAR and hPXR by atorvastatin and the subsequent induction of not only CYP2B6 and CYP3A4 but also of CYP2C9 present an additional target by which atorvastatin, a widely used cholesterol-lowering drug, can modify the kinetics of numerous drugs. Atorvastatin 160-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 18971317-10 2009 Activation of hCAR and hPXR by atorvastatin and the subsequent induction of not only CYP2B6 and CYP3A4 but also of CYP2C9 present an additional target by which atorvastatin, a widely used cholesterol-lowering drug, can modify the kinetics of numerous drugs. Cholesterol 188-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 19077919-3 2009 RESULTS: In the early phase of warfarin therapy, the combined genotypes of CYP2C9 and VKORC1 caused statistically significant difference in warfarin dose from day 7 of warfarin dosing and the subsequent time course of dose increase showed significant difference among the three different genotypes (P<0.001). Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19033450-1 2009 In vitro data on the metabolism of the antifungal voriconazole suggest that its pharmacokinetics might be influenced by the activity of CYP2C19, CYP2C9, and CYP3A. Voriconazole 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 19077919-1 2009 OBJECTIVES: The effect of CYP2C9 and vitamin K epoxide reductase complex subunit 1 (VKORC1) genotypes was evaluated for the early-phase and steady-state warfarin dosing in Korean patients with mechanical heart valve replacement. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 18541345-0 2009 The metabolism of CYP2C9 and CYP2C19 for gliclazide by homology modeling and docking study. Gliclazide 41-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 18541345-5 2009 Then, with the refined model of CYP2C19 and the crystal structure of CYP2C9, the metabolisms of them for gliclazide in two different metabolic pathways were studied and the results show that both enzymes have more favorable interaction energies and stronger affinity with gliclazide in methylhydroxylation pathway than in 6beta-hydroxylation pathway. Gliclazide 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 18541345-5 2009 Then, with the refined model of CYP2C19 and the crystal structure of CYP2C9, the metabolisms of them for gliclazide in two different metabolic pathways were studied and the results show that both enzymes have more favorable interaction energies and stronger affinity with gliclazide in methylhydroxylation pathway than in 6beta-hydroxylation pathway. Gliclazide 272-282 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 18541345-6 2009 It is exciting that substrate inhibition phenomenon can be found in metabolisms of CYP2C9 and CYP2C19 for gliclazide in two metabolic pathways. Gliclazide 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 18541345-7 2009 Gliclazide can change the conformation of the active sites and decrease obviously the affinities between gliclazide in the active site and enzymes when it is docked in the second active sites in CYP2C9 and CYP2C19. Gliclazide 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 18541345-7 2009 Gliclazide can change the conformation of the active sites and decrease obviously the affinities between gliclazide in the active site and enzymes when it is docked in the second active sites in CYP2C9 and CYP2C19. Gliclazide 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 19179293-5 2009 A substantial number of studies demonstrate that common variants of two genes, VKORC1 and CYP2C9, along with other nongenetic factors, correlate significantly with warfarin dosing. Warfarin 164-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 19179293-6 2009 The genotypes of VKORC1 and CYP2C9 alone account for nearly 3 times more of the variability ( approximately 30%) in warfarin dosing than do age, weight, gender, and other clinical factors combined ( approximately 12%). Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 19077919-3 2009 RESULTS: In the early phase of warfarin therapy, the combined genotypes of CYP2C9 and VKORC1 caused statistically significant difference in warfarin dose from day 7 of warfarin dosing and the subsequent time course of dose increase showed significant difference among the three different genotypes (P<0.001). Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19077919-3 2009 RESULTS: In the early phase of warfarin therapy, the combined genotypes of CYP2C9 and VKORC1 caused statistically significant difference in warfarin dose from day 7 of warfarin dosing and the subsequent time course of dose increase showed significant difference among the three different genotypes (P<0.001). Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19077919-6 2009 The observed warfarin maintenance dose was best explained by a model including covariates of age, weight, concurrent congestive heart failure/cardiomyopathy, INR-increasing drugs, aspirin, dietary supplements, and CYP2C9 and VKORC1 genotypes (R=0.56). Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 19077919-7 2009 CONCLUSION: The heterozygous CYP2C9 and VKORC1 genotypes influence warfarin dosing in an early phase as well as steady state of warfarin therapy in Korean patients with mechanical heart valve replacement. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19077919-7 2009 CONCLUSION: The heterozygous CYP2C9 and VKORC1 genotypes influence warfarin dosing in an early phase as well as steady state of warfarin therapy in Korean patients with mechanical heart valve replacement. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19139476-4 2009 Cytochrome P-450 isoenzyme 2C9 (CYP2C9) metabolizes S-warfarin into two inactive metabolites. Warfarin 52-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-30 18574025-1 2009 Genetic variants of cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) are known to influence warfarin dose, but the effect of other genes has not been fully elucidated. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-39 18574025-1 2009 Genetic variants of cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) are known to influence warfarin dose, but the effect of other genes has not been fully elucidated. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18574025-3 2009 CYP2C9*2 and *3 explained 12% (P = 6.63 x 10(-34)) of the variation in warfarin dose, while a single VKORC1 SNP explained 30% (P = 9.82 x 10(-100)). Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18574025-7 2009 A multiple regression model using the predictors CYP2C9, VKORC1, age, sex, and druginteractions explained 59% of the variance in warfarin dose, and 53% in an independent sample of 181 Swedish individuals. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 18574025-8 2009 In conclusion, CYP2C9 and VKORC1 significantly influenced warfarin dose and predicted individuals predisposed to unstable anticoagulation. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 19348697-3 2009 Genotype for CYP2C9, which encodes the main cytochrome P450 enzyme that metabolizes warfarin, and VKORC1, the gene encoding the warfarin target vitamin K epoxide reductase, together account for approximately 30% of the variability in dose requirement. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19348697-7 2009 A recently published whole genome association study confirms that the main genes important in warfarin response are CYP2C9 and VKORC1. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 19139476-4 2009 Cytochrome P-450 isoenzyme 2C9 (CYP2C9) metabolizes S-warfarin into two inactive metabolites. Warfarin 52-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 19139476-5 2009 Genetic variations to the gene encoding CYP2C9 (CYP2C9 ) are known to affect warfarin clearance. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 19139476-5 2009 Genetic variations to the gene encoding CYP2C9 (CYP2C9 ) are known to affect warfarin clearance. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 19117406-0 2009 SYBR Green-based real-time PCR assay for detection of VKORC1 and CYP2C9 polymorphisms that modulate warfarin dose requirement. sybr green 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 26110197-6 2009 CYP2C9 is the principal cytochrome P450 enzyme that modulates the anticoagulant activity of warfarin. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26110197-7 2009 From results of clinical studies, genetic variation in the CYP2C9 and/or VKORC1 genes can, in concert with clinical factors, predict how each individual responds to warfarin. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 26110197-11 2009 Pharmacogenomic testing of CYP2C9 or VKORC1 alleles to predict a patient"s response to warfarin occurs ideally prior to initiation of the drug. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 19067473-4 2009 Based on in vitro studies, dexlansoprazole has the potential to inhibit activity of these isoenzymes and also may induce human hepatic CYP1A and CYP2C9 activity. Dexlansoprazole 27-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 19117406-0 2009 SYBR Green-based real-time PCR assay for detection of VKORC1 and CYP2C9 polymorphisms that modulate warfarin dose requirement. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19117406-1 2009 BACKGROUND: Polymorphisms in VKORC1 (vitamin K epoxide reductase complex subunit 1) and CYP2C9 (cytochrome P450 2C9) genes are considered the major genetic factors modulating warfarin dose requirement. Warfarin 175-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19397481-5 2009 Growing evidence indicates that up to 60% of the individual pharmacological response to coumarins might be due to genetic variables and affected by polymorphisms in the genes encoding two enzymes, namely, vitamin K epoxide reductase (VKOR) and cytochrome P450 CYP2C9. Coumarins 88-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 260-266 19117406-1 2009 BACKGROUND: Polymorphisms in VKORC1 (vitamin K epoxide reductase complex subunit 1) and CYP2C9 (cytochrome P450 2C9) genes are considered the major genetic factors modulating warfarin dose requirement. Warfarin 175-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-115 19117406-3 2009 METHODS: In this study, we developed and validated a melting temperature (T(m))-shift genotyping assay for detection of VKORC1 C1173T and CYP2C9 A1075C polymorphisms by using SYBR Green-based multiplex allele-specific PCR and consequent melting curve analysis. sybr green 175-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 19071883-9 2009 In addition to the clinical pharmacokinetic analysis, the ability of mipomersen sodium to inhibit the major CYP isoform enzymes (namely CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4) was evaluated in cryo-preserved human hepatocytes in vitro. Sodium 80-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 19725591-6 2009 Etravirine is metabolized by cytochrome P450 (CYP) 3A, 2C9 and 2C19; the metabolites are subsequently glucuronidated by uridine diphosphate glucuronosyltransferase. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-58 19725591-8 2009 Etravirine has the potential for interactions by inducing CYP3A and inhibiting CYP2C9 and 2C19; it is a mild inhibitor of P-glycoprotein but not a substrate. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18948380-10 2009 Human P450 isoforms that showed activity toward multiple pyrethroids were CYP2C8, CYP2C9, CYP2C19, and CYP3A4. Pyrethrins 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 22461099-8 2009 However, only variations in VKORC1 and CYP2C9 have consistently been associated with drug response (coumarins) and have clinical implications. Coumarins 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Tolbutamide 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Glyburide 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Diclofenac 86-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Celecoxib 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Torsemide 109-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). phenytoin losartan 121-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-2 2009 CYP2C9 metabolizes more than 100 therapeutic drugs, including tolbutamide, glyburide, diclofenac, celecoxib, torasemide, phenytoin losartan, and S-warfarin). Warfarin 145-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-4 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Steroids 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-4 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Melatonin 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-4 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Retinoids 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-4 2009 CYP2C9 also plays a role in the metabolism of several endogenous compounds such as steroids, melatonin, retinoids and arachidonic acid. Arachidonic Acid 118-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-9 2009 CYP2C9 is activated by dapsone and its analogues and R-lansoprazole in a stereo-specific and substrate-dependent manner, probably through binding to the active site and inducing positive cooperativity. Dapsone 23-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-9 2009 CYP2C9 is activated by dapsone and its analogues and R-lansoprazole in a stereo-specific and substrate-dependent manner, probably through binding to the active site and inducing positive cooperativity. Dexlansoprazole 53-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Rifampin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Rifampin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Phenobarbital 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Phenobarbital 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Dexamethasone 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19515014-10 2009 CYP2C9 is subject to induction by rifampin, phenobarbital, and dexamethasone, indicating the involvement of pregnane X receptor, constitutive androstane receptor and glucocorticoid receptor in the regulation of CYP2C9. Dexamethasone 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 19515014-12 2009 Tienilic acid, suprofen and silybin are mechanism-based inhibitors of CYP2C9. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 19515014-12 2009 Tienilic acid, suprofen and silybin are mechanism-based inhibitors of CYP2C9. Suprofen 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 19515014-12 2009 Tienilic acid, suprofen and silybin are mechanism-based inhibitors of CYP2C9. Silybin 28-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 19354002-0 2009 Rosuvastatin-induced rhabdomyolysis probably via CYP2C9 saturation. Rosuvastatin Calcium 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Rosuvastatin Calcium 23-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Rosuvastatin Calcium 23-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Telmisartan 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Telmisartan 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Rosuvastatin Calcium 131-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 19354002-8 2009 Our patient was taking rosuvastatin, warfarin and telmisartan, which are metabolised by CYP2C9; we therefore hypothesised that the rosuvastatin-induced rhabdomyolysis was probably by CYP2C9 enzyme saturation. Rosuvastatin Calcium 131-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 19067731-8 2009 Heterogeneity in the way patients respond to aspirin and clopidogrel may in part reflect variation in cyclooxygenase (COX)-1, COX-2, glycoprotein (GP) Ib alpha, GP Ia/IIa, GP IIb/IIIa, UGT1A6*2, P2Y(1), P2Y(12), CYP2C9, CYP3A4 and CYP3A5 genotypes. Aspirin 45-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 19067731-8 2009 Heterogeneity in the way patients respond to aspirin and clopidogrel may in part reflect variation in cyclooxygenase (COX)-1, COX-2, glycoprotein (GP) Ib alpha, GP Ia/IIa, GP IIb/IIIa, UGT1A6*2, P2Y(1), P2Y(12), CYP2C9, CYP3A4 and CYP3A5 genotypes. Clopidogrel 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 19745563-8 2009 The percent variability explained by VKORC1 diplotype status was 59.1% while the CYP2C9 genotype status accounted for 6.9% variability in warfarin dose requirements. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 20408500-2 2009 Inhibition of CYP1A2, CYP2E1, CYP2C19, CYP2C9, CYP2D6, and CYP3A4 by CGS was assessed using recombinant human enzymes incubated with CGS (up to 3 mM expressed as free base). cysteinylglycine 69-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 20408500-3 2009 Induction of CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP3A4 by CGS (0.01, 0.3 and 3 mM) was evaluated in cryopreserved human hepatocytes, by determining CYP mRNA expression using quantitative RT-PCR. cysteinylglycine 59-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 19053752-0 2008 Cytochrome P450 2C9 type II binding studies on quinoline-4-carboxamide analogues. quinoline-4-carboxamide 47-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 18838506-0 2009 Mechanism-based inactivation of cytochrome P450 2C9 by tienilic acid and (+/-)-suprofen: a comparison of kinetics and probe substrate selection. Ticrynafen 55-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 18838506-0 2009 Mechanism-based inactivation of cytochrome P450 2C9 by tienilic acid and (+/-)-suprofen: a comparison of kinetics and probe substrate selection. Suprofen 73-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 23226035-7 2009 For example, it has been found that patients with CYP2C9 and VKORC1 gene variations have a different response to warfarin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 18754597-0 2008 QM/MM modeling of benzene hydroxylation in human cytochrome P450 2C9. Benzene 18-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-68 18754597-1 2008 The mechanism of benzene hydroxylation was investigated in the realistic enzyme environment of the human CYP 2C9 by using quantum mechanical/molecular mechanical (QM/MM) calculations of the whole reaction profile using the B3LYP method to describe the QM region. Benzene 17-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-112 19430171-2 2009 In this study, we investigated whether these mice show similar metabolic profile to humans by examining the hydroxylation of S-warfarin reported to be mainly metabolized to S-7-hydroxywarfarin (7-OH-warfarin), catalyzed by CYP2C9, in humans. Warfarin 125-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-229 19430171-2 2009 In this study, we investigated whether these mice show similar metabolic profile to humans by examining the hydroxylation of S-warfarin reported to be mainly metabolized to S-7-hydroxywarfarin (7-OH-warfarin), catalyzed by CYP2C9, in humans. 7-hydroxywarfarin 173-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-229 19430171-2 2009 In this study, we investigated whether these mice show similar metabolic profile to humans by examining the hydroxylation of S-warfarin reported to be mainly metabolized to S-7-hydroxywarfarin (7-OH-warfarin), catalyzed by CYP2C9, in humans. 7-oh-warfarin 194-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-229 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Clotrimazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Rifampin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Ritonavir 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Phenytoin 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-3 2009 Clotrimazole, rifampin, ritonavir, phenytoin, and phenobarbital induced CYP2C9 consistent with previous findings for CYP3A4. Phenobarbital 50-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 19202563-6 2009 In 17 donors, rifampin increased mean basal CYP2C9 activity from 59 +/- 43 to 143 +/- 68 pmol/mg protein/min; fold induction ranged from 1.4- to 6.4-fold. Rifampin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 18992346-0 2009 Pharmacogenetic relevance of the CYP2C9*3 allele in a tenoxicam bioequivalence study performed on Spaniards. tenoxicam 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 18992346-1 2009 We performed a study to quantify CYP2C9 and CYP2C8 alleles influence on the variability observed in tenoxicam pharmacokinetic (PK) and implication in a bioequivalence study design performed on Spaniards. tenoxicam 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 18992346-5 2009 CYP2C9*3 allele is associated to a longer elimination time of tenoxicam. tenoxicam 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19219744-4 2009 This method was used to determine the bioactivation of acetaminophen at two concentrations: 50 microM therapeutic and 1 mM toxic by using nine human recombinant CYP enzymes: CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4; and with different microsomes from experimental animals. Acetaminophen 55-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 19017829-1 2008 OBJECTIVE: To report a case in which the anticoagulation effect of warfarin appeared to be potentiated by torsemide, possibly due to an interference of metabolism through competition for the CYP2C9 isoenzyme and protein-binding displacement of warfarin. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 19026171-0 2008 Determination of the inhibitory potential of 6 fluoroquinolones on CYP1A2 and CYP2C9 in human liver microsomes. 6 fluoroquinolones 45-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 19026171-2 2008 METHODS: Probe substrates, phenacetin (CYP1A2), and tolbutamide (CYP2C9) were incubated with human liver microsomes and the metabolites were analyzed by liquid chromatography/mass spectrometry using electrospray ionization in positive or negative mode. Tolbutamide 52-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19026171-4 2008 The inhibitory potential of fluoroquinolones on CYP1A2 and CYP2C9 was investigated. Fluoroquinolones 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 19026171-6 2008 Ciprofloxacin showed weak inhibition on both the activity of CYP1A2 (IC50 135 micromol/L) and CYP2C9 (IC50 180 micromol/L), whereas levofloxacin inhibited only CYP2C9 (IC50 210 micromol/L). Ciprofloxacin 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 19017829-1 2008 OBJECTIVE: To report a case in which the anticoagulation effect of warfarin appeared to be potentiated by torsemide, possibly due to an interference of metabolism through competition for the CYP2C9 isoenzyme and protein-binding displacement of warfarin. Torsemide 106-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 19026171-8 2008 CONCLUSION: Caderofloxacin, antofloxacin, moxifloxacin, and gatifloxacin are negligible inhibitors to CYP1A2 and CYP2C9. cadrofloxacin 12-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 19017829-5 2008 DISCUSSION: Both torsemide and warfarin are highly protein-bound to albumin and are major substrates for the CYP2C9 isoenzyme. Torsemide 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 19017829-5 2008 DISCUSSION: Both torsemide and warfarin are highly protein-bound to albumin and are major substrates for the CYP2C9 isoenzyme. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 19026171-8 2008 CONCLUSION: Caderofloxacin, antofloxacin, moxifloxacin, and gatifloxacin are negligible inhibitors to CYP1A2 and CYP2C9. antofloxacin 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 19026171-8 2008 CONCLUSION: Caderofloxacin, antofloxacin, moxifloxacin, and gatifloxacin are negligible inhibitors to CYP1A2 and CYP2C9. Moxifloxacin 42-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 19017829-8 2008 Therefore, it is possible that the addition of torsemide may potentiate the anticoagulant effect of warfarin by (1) competition for metabolism through CYP2C9, with a decrease in the clearance of warfarin, and (2) protein-binding displacement of warfarin from albumin, transiently potentiating anticoagulant activity. Torsemide 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 19026171-8 2008 CONCLUSION: Caderofloxacin, antofloxacin, moxifloxacin, and gatifloxacin are negligible inhibitors to CYP1A2 and CYP2C9. Gatifloxacin 60-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 19017829-8 2008 Therefore, it is possible that the addition of torsemide may potentiate the anticoagulant effect of warfarin by (1) competition for metabolism through CYP2C9, with a decrease in the clearance of warfarin, and (2) protein-binding displacement of warfarin from albumin, transiently potentiating anticoagulant activity. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 18787056-4 2008 In the presence of HLMs, sulfaphenazole (CYP2C9 inhibitor), and anti-CYP2C9 monoclonal antibodies (mAbs) inhibited (73-100%) the 2- and 3-hydroxylation of both IBU enantiomers (1 and 20 microM). Sulfaphenazole 25-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18765684-1 2008 Sulfamethoxazole is metabolized by microsomal CYP2C9 to a hydroxylamine that is thought to be responsible for the relatively high incidence of hypersensitivity reactions associated with the drug. Hydroxylamine 58-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 18765684-1 2008 Sulfamethoxazole is metabolized by microsomal CYP2C9 to a hydroxylamine that is thought to be responsible for the relatively high incidence of hypersensitivity reactions associated with the drug. Sulfamethoxazole 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 18799803-3 2008 SKF525A and its metabolite and primary amine analog all inhibited CYP2B6-, CYP2C9-, CYP2C19-, CYP2D6-, and CYP3A-selective reactions to varying degrees but had little effect on CYP1A2, CYP2A6, and CYP2E1 reactions. Proadifen 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 18784266-0 2008 CYP2C9-catalyzed metabolism of S-warfarin to 7-hydroxywarfarin in vivo and in vitro in chimeric mice with humanized liver. Warfarin 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18784266-0 2008 CYP2C9-catalyzed metabolism of S-warfarin to 7-hydroxywarfarin in vivo and in vitro in chimeric mice with humanized liver. 7-hydroxywarfarin 45-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18799803-3 2008 SKF525A and its metabolite and primary amine analog all inhibited CYP2B6-, CYP2C9-, CYP2C19-, CYP2D6-, and CYP3A-selective reactions to varying degrees but had little effect on CYP1A2, CYP2A6, and CYP2E1 reactions. Amines 39-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 18787056-4 2008 In the presence of HLMs, sulfaphenazole (CYP2C9 inhibitor), and anti-CYP2C9 monoclonal antibodies (mAbs) inhibited (73-100%) the 2- and 3-hydroxylation of both IBU enantiomers (1 and 20 microM). Ibuprofen 160-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18787056-4 2008 In the presence of HLMs, sulfaphenazole (CYP2C9 inhibitor), and anti-CYP2C9 monoclonal antibodies (mAbs) inhibited (73-100%) the 2- and 3-hydroxylation of both IBU enantiomers (1 and 20 microM). Ibuprofen 160-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 18784266-2 2008 In this study, we investigated whether these mice have a capacity for drug metabolism similar to that of humans by examining hydroxylation of S-warfarin, which is predominantly metabolized to S-7-hydroxywarfarin, catalyzed by CYP2C9, in humans but not mice. Warfarin 142-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 226-232 18784266-2 2008 In this study, we investigated whether these mice have a capacity for drug metabolism similar to that of humans by examining hydroxylation of S-warfarin, which is predominantly metabolized to S-7-hydroxywarfarin, catalyzed by CYP2C9, in humans but not mice. 7-hydroxywarfarin 192-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 226-232 18787056-7 2008 When (S)-(+)-IBU and (R)-(-)-IBU (1 microM) were incubated with a panel of recombinant human P450s, only CYP2C9 formed appreciable amounts of the hydroxy metabolites. Ibuprofen 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 18787056-7 2008 When (S)-(+)-IBU and (R)-(-)-IBU (1 microM) were incubated with a panel of recombinant human P450s, only CYP2C9 formed appreciable amounts of the hydroxy metabolites. Ibuprofen 21-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 18787056-11 2008 On the other hand, CYP2C9 inhibition and genotype are expected to have an impact on the PK of (S)-(+)-IBU. Ibuprofen 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 25855835-20 2008 CONCLUSIONS: Certain CYP2C9 and VKORC1 variants are associated with lower warfarin maintenance doses, and CYP2C9 variants are associated with increased bleeding rates among patients who use warfarin. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 25855835-6 2008 RESULTS: The 99 included articles reported 103 studies: 29 tested the association of CYP2C9 and the response to warfarin; 19 tested the association of VKORC1 and the response to warfarin; 44 tested the association of Apo E and the response to statins; and 11 tested the association of MTHFR with the response to antifolate chemotherapy.CYP2C9 AND VKORC1 GENE POLYMORPHISMS AND RESPONSE TO WARFARIN THERAPY: Of the 29 studies of CYP2C9 gene polymorphisms, 26 evaluated their association with responses to maintenance does of warfarin. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 19132230-7 2008 The impact of the CYP2C9 and VKORC1 genotype is most significant during the initial period of coumarin anticoagulant therapy. coumarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 19132230-9 2008 The CYP2C9 polymorphism is associated with delayed stabilisation for coumarin anticoagulants. coumarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 18922023-0 2008 Functional analysis of phenylalanine residues in the active site of cytochrome P450 2C9. Phenylalanine 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-87 18922023-1 2008 The two published crystal structures of cytochrome P450 2C9, complexed with ( S)-warfarin or flurbiprofen, implicate a cluster of three active site phenylalanine residues (F100, F114, F476) in ligand binding. Warfarin 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-59 18922023-1 2008 The two published crystal structures of cytochrome P450 2C9, complexed with ( S)-warfarin or flurbiprofen, implicate a cluster of three active site phenylalanine residues (F100, F114, F476) in ligand binding. Flurbiprofen 93-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-59 18922023-1 2008 The two published crystal structures of cytochrome P450 2C9, complexed with ( S)-warfarin or flurbiprofen, implicate a cluster of three active site phenylalanine residues (F100, F114, F476) in ligand binding. Phenylalanine 148-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-59 18922023-3 2008 To elucidate the importance of CYP2C9"s active site phenylalanines on substrate binding, orientation, and catalytic turnover, a series of leucine and tryptophan mutants were constructed and their interactions with ( S)-warfarin and ( S)-flurbiprofen examined. Phenylalanine 52-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18922023-3 2008 To elucidate the importance of CYP2C9"s active site phenylalanines on substrate binding, orientation, and catalytic turnover, a series of leucine and tryptophan mutants were constructed and their interactions with ( S)-warfarin and ( S)-flurbiprofen examined. Leucine 138-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18922023-3 2008 To elucidate the importance of CYP2C9"s active site phenylalanines on substrate binding, orientation, and catalytic turnover, a series of leucine and tryptophan mutants were constructed and their interactions with ( S)-warfarin and ( S)-flurbiprofen examined. Tryptophan 150-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18922023-3 2008 To elucidate the importance of CYP2C9"s active site phenylalanines on substrate binding, orientation, and catalytic turnover, a series of leucine and tryptophan mutants were constructed and their interactions with ( S)-warfarin and ( S)-flurbiprofen examined. Warfarin 214-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18922023-3 2008 To elucidate the importance of CYP2C9"s active site phenylalanines on substrate binding, orientation, and catalytic turnover, a series of leucine and tryptophan mutants were constructed and their interactions with ( S)-warfarin and ( S)-flurbiprofen examined. Flurbiprofen 232-249 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 25855835-8 2008 Carriers of the CYP2C9 gene variant alleles *2 or *3 had lower mean maintenance warfarin dose requirements than did non-carriers. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25855835-9 2008 Few studies investigated the relationship between genetic variations in CYP2C9 or VKORC1 and warfarin dose requirements in the induction phase. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 25855835-10 2008 CYP2C9 variants were associated with an increased rate of bleeding complications during the induction phase of warfarin therapy, but the studies did not report whether affected patients had normal or supratherapeutic INR ranges. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25855835-20 2008 CONCLUSIONS: Certain CYP2C9 and VKORC1 variants are associated with lower warfarin maintenance doses, and CYP2C9 variants are associated with increased bleeding rates among patients who use warfarin. Warfarin 190-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 19018719-0 2008 VKORC1 and CYP2C9 allelic variants influence acenocoumarol dose requirements in Greek patients. Acenocoumarol 45-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 18787075-5 2008 Moreover, VEGF-stimulated endothelial cell sprouting in a modified spheroid assay was reduced by CYP2C antisense oligonucleotides. Oligonucleotides 113-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-102 19069171-4 2008 Recently genetic factors such as the CYP2C9 and VKORC1 genes have been demonstrated to be determinants of warfarin response. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19069171-5 2008 CYP2C9 is the enzyme primarily responsible for the metabolic clearance of the S-enantiomer of warfarin. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 19069171-8 2008 On the basis of these observations, the Food and Drug Administration (FDA) approved a labeling change for warfarin that includes the genetic information of VKORC1 and CYP2C9 as factors influencing interindividual variability in warfarin dosing. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 19069171-8 2008 On the basis of these observations, the Food and Drug Administration (FDA) approved a labeling change for warfarin that includes the genetic information of VKORC1 and CYP2C9 as factors influencing interindividual variability in warfarin dosing. Warfarin 228-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 19108785-11 2008 Nilotinib is a competitive inhibitor of cytochrome P450 (CYP) 3A4, CYP2C8, CYP2C9, and CYP2D6. nilotinib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 19211991-0 2008 Limited applicability of 7-methoxy-4-trifluoromethylcoumarin as a CYP2C9-selective substrate. 7-methoxy-4-trifluoromethylcoumarin 25-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 19211991-2 2008 7-Methoxy-4-trifluoromethylcoumarin (MFC) has been reported as a CYP2C9-selective substrate. 7-methoxy-4-trifluoromethylcoumarin 0-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 19211991-2 2008 7-Methoxy-4-trifluoromethylcoumarin (MFC) has been reported as a CYP2C9-selective substrate. 7-methoxy-4-trifluoromethylcoumarin 37-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17961958-3 2008 To our knowledge, this is the first description of phenytoin intoxication due to CYP2C9 inhibition by clozapine. Phenytoin 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 17961958-3 2008 To our knowledge, this is the first description of phenytoin intoxication due to CYP2C9 inhibition by clozapine. Clozapine 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 19018719-8 2008 In a multivariable regression model, CYP2C9, VKORC1 genotypes and age explained 55% of acenocoumarol dosing variability. Acenocoumarol 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 19018719-9 2008 CONCLUSION: VKORC1-1639G>A, CYP2C9*2 and CYP2C9*3 polymorphisms were found to predispose to acenocoumarol sensitivity in Greeks. Acenocoumarol 95-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 19018719-9 2008 CONCLUSION: VKORC1-1639G>A, CYP2C9*2 and CYP2C9*3 polymorphisms were found to predispose to acenocoumarol sensitivity in Greeks. Acenocoumarol 95-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 18548238-2 2008 We have demonstrated the relevance of CYP2C9 genotypes for diclofenac 4"-hydroxylation in healthy volunteers, so that the present study was aimed at analyzing the role of both CYP2C8 and CYP2C9 genotypes on diclofenac metabolism, as well as determining the CYP2C8 allele frequencies and their relationship with CYP2C9 variants. diclofenac 4" 59-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 18680736-7 2008 The associations of SNPs rs9934438 and rs9923231 of VKORC1, the 3 (rs1057910) and C(-65) (rs9332127) alleles of CYP2C9, and SNP rs4653436 of EPHXI with the dose of warfarin were significant. Warfarin 164-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 18680736-8 2008 CONCLUSION: A multiple regression model based on the genetic polymorphisms of VKORC1, CYP2C9, EPHX1 and the non-genetic factors of age and body weight can explain 40.2% of the variance in warfarin dose in Han Chinese patients. Warfarin 188-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 18758764-3 2008 Carriers of cytochrome P450 2C9 (CYP2C9)-deficient alleles have a reduced clearance of warfarin and are exposed to dramatic overdoses in the first weeks of treatment. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-31 18758764-3 2008 Carriers of cytochrome P450 2C9 (CYP2C9)-deficient alleles have a reduced clearance of warfarin and are exposed to dramatic overdoses in the first weeks of treatment. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 18758764-6 2008 Several warfarin dosing algorithms have been constructed, adapted on CYP2C9 and VKORC1 genotypes and clinical factors, to predict the best dose for each patient. Warfarin 8-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 18669935-3 2008 We hypothesized that inhibition of CYP 2C9 signaling with sulfaphenazole would improve EDD in older, but not young, healthy sedentary adults. Sulfaphenazole 58-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-42 18606741-4 2008 We expressed and purified the R108E and R108E/D293N mutants and compared their ability with that of native CYP2C9 to interact with (S)-warfarin, diclofenac, pyrene, propranolol, and ibuprofen amine. Warfarin 131-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 18606741-4 2008 We expressed and purified the R108E and R108E/D293N mutants and compared their ability with that of native CYP2C9 to interact with (S)-warfarin, diclofenac, pyrene, propranolol, and ibuprofen amine. Diclofenac 145-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 18606741-4 2008 We expressed and purified the R108E and R108E/D293N mutants and compared their ability with that of native CYP2C9 to interact with (S)-warfarin, diclofenac, pyrene, propranolol, and ibuprofen amine. pyrene 157-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 18606741-4 2008 We expressed and purified the R108E and R108E/D293N mutants and compared their ability with that of native CYP2C9 to interact with (S)-warfarin, diclofenac, pyrene, propranolol, and ibuprofen amine. Propranolol 165-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 18679669-2 2008 In vitro, nebicapone has showed an inhibitory effect upon CYP2C9, which is responsible for the metabolism of S-warfarin. nebicapone 10-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 18679669-2 2008 In vitro, nebicapone has showed an inhibitory effect upon CYP2C9, which is responsible for the metabolism of S-warfarin. Warfarin 109-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 18548238-2 2008 We have demonstrated the relevance of CYP2C9 genotypes for diclofenac 4"-hydroxylation in healthy volunteers, so that the present study was aimed at analyzing the role of both CYP2C8 and CYP2C9 genotypes on diclofenac metabolism, as well as determining the CYP2C8 allele frequencies and their relationship with CYP2C9 variants. Diclofenac 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 18975556-7 2008 Moreover, testing for CYP2C9*3 and VKORC1 6484 C>T was shown to be useful for the safe administration of warfarin. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 18855533-6 2008 RESULTS: VKORC1 explained 18% (30% with CYP2C9) variability in warfarin dose among European-Americans and 5% (8% with CYP2C9) among African-Americans. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 18855533-10 2008 CONCLUSIONS: Our findings support the use of CYP2C9, VKORC1 polymorphisms (rs9934438 or rs9923231) and clinical covariates to predict warfarin dose in both African- and European-Americans. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 18855533-11 2008 A uniform set of common polymorphisms in CYP2C9 and VKORC1, and limited clinical covariates can be used to improve warfarin dose prediction for a racially diverse population. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18584988-3 2008 The 384-well assay used human liver microsomes in conjunction with a cocktail of probe substrates metabolized by the five major CYPs (tacrine for CYP1A2, diclofenac for CYP2C9, (S)-mephenytoin for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4). Diclofenac 154-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 18682545-12 2008 CONCLUSIONS: There is growing evidence that variant alleles for CYP2C9 and VKORC1 genotypes account for a proportion of the variability seen in warfarin dosing. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 18585438-3 2008 CYP2C9 and CYP2C19 possessed high enantioselectivity favoring the formation of S-Mono-OH-M; CYP2C3 showed no enantioselectivity, whereas CYP2C5 slightly favored the formation of R-Mono-OH-M. 1,1,1-trichloro-2-(4-hydroxyphenyl)-2-(4-methoxyphenyl)ethane 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18585438-3 2008 CYP2C9 and CYP2C19 possessed high enantioselectivity favoring the formation of S-Mono-OH-M; CYP2C3 showed no enantioselectivity, whereas CYP2C5 slightly favored the formation of R-Mono-OH-M. r-mono-oh-m 178-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18542936-0 2008 VKORC1 and CYP2C9 polymorphisms are associated with warfarin dose requirements in Turkish patients. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 18542936-1 2008 OBJECTIVES: The objective of this study was to determine the quantitative influence of vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP 2C9) polymorphisms on warfarin dose requirements in Turkish patients. Warfarin 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-165 18542936-6 2008 Patients who carried VKORC1 and CYP2C9 variants needed a 40% lower mean weekly warfarin dose compared to wild types. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 18542936-8 2008 CONCLUSION: Polymorphisms in VKORC1 and CYP2C9 genes were important determinants of warfarin dose requirements in Turkish patients. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 18752379-2 2008 We review the evidence of the influence of the two key genes of interest, the cytochrome P450 2C9 gene, CYP2C9, and the vitamin K epoxide reductase complex 1 gene, VKORC1, on warfarin response and discuss the implications of current knowledge for clinical practice. Warfarin 175-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-97 18752379-2 2008 We review the evidence of the influence of the two key genes of interest, the cytochrome P450 2C9 gene, CYP2C9, and the vitamin K epoxide reductase complex 1 gene, VKORC1, on warfarin response and discuss the implications of current knowledge for clinical practice. Warfarin 175-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 18752379-3 2008 The influence of CYP2C9 and VKORC1 genotypes on warfarin dose requirements has been consistently demonstrated in diverse racial and ethnic patient groups in observational studies and randomized clinical trials. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 18480186-3 2008 The curcuminoid extract inhibited SULT > CYP2C19 > CYP2B6 > UGT > CYP2C9 > CYP3A activities with IC(50) values ranging from 0.99 +/- 0.04 to 25.3 +/- 1.3 microM, whereas CYP2D6, CYP1A2, and CYP2E1 activities were less affected (IC(50) values > 60 microM). curcuminoid 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 18535201-1 2008 Warfarin dosing is correlated with polymorphisms in vitamin K epoxide reductase complex 1 (VKORC1) and the cytochrome P450 2C9 (CYP2C9) genes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-126 18535201-1 2008 Warfarin dosing is correlated with polymorphisms in vitamin K epoxide reductase complex 1 (VKORC1) and the cytochrome P450 2C9 (CYP2C9) genes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 18763667-7 2008 The combination of physiologic factors (30%), CYP2C9 variations (20%) and VKORC1 variants (25%) accounts for approximately 75% of warfarin dose variability. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 19032008-5 2008 Following completion of the Human Genome Project, several genetic variants of CYP2C9 and VKORC1 have been identified that account for a greater proportion of the variability in patient response to warfarin than is explained by nongenetic factors. Warfarin 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 18781859-3 2008 For example, persons of African descent have lower allele frequencies of the CYP2C9*2 and *3 and VKORC1 1173T allele, which have been associated with lower warfarin dose requirements in Caucasians. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 18816302-0 2008 Disposition of a CYP2C9 phenotyping agent, losartan, is not influenced by the common 3435C > T variation of the drug transporter gene ABCB1 (MDR1). Losartan 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-52 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. losartan carboxylic acid 24-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-52 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. losartan carboxylic acid 24-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 18816302-1 2008 Losartan is oxidized to E3174 by cytochrome P450 2C9 (CYP2C9); it has been suggested as a useful probe drug for CYP2C9 activity. losartan carboxylic acid 24-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). Clopidogrel 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 301-307 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). Ticlopidine 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 301-307 18474675-2 2008 Mechanism-based CYP2B6 inhibitors (i.e., clopidogrel, ticlopidine, and triethylenethiophoramide) significantly inhibited the formation of M1 from sibutramine and M2 from M1, respectively; in contrast, no effect was observed when using potent inhibitors of eight P450 isozymes (CYP1A2, CYP2A6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A). triethylenethiophoramide 71-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 301-307 18480186-8 2008 All three curcuminoids had similar effects on CYP3A, UGT, and SULT activity, but demethoxycurcumin (IC(50) = 8.8 +/- 1.2 microM) was more active against CYP2C9 than either curcumin or bisdemethoxycurcumin (IC(50) > 50 microM). demethoxycurcumin 81-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 18480186-10 2008 administered curcuminoid/piperine combination is most likely to inhibit CYP3A, CYP2C9, UGT, and SULT metabolism within the intestinal mucosa. curcuminoid 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18480186-10 2008 administered curcuminoid/piperine combination is most likely to inhibit CYP3A, CYP2C9, UGT, and SULT metabolism within the intestinal mucosa. piperine 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18971529-0 2008 Genetic variations of CYP2C9 in 724 Japanese individuals and their impact on the antihypertensive effects of losartan. Losartan 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 18662109-7 2008 Etravirine is an inducer of CYP3A4 and a weak inhibitor of CYP2C9, CYP2C19 and P-glycoprotein. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 18971529-1 2008 CYP2C9, a drug-metabolizing enzyme, converts the angiotensin II receptor blocker losartan to its active form, which is responsible for its antihypertensive effect. Losartan 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18971529-5 2008 After 3 months of losartan treatment, systolic blood pressure was not lowered in two patients with CYP2C9* 1/*30, suggesting that they exhibited impaired in vivo CYP2C9 activity. Losartan 18-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 18971529-6 2008 CYP2C9*30 might be associated with a diminished response to the antihypertensive effects of losartan. Losartan 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18485885-0 2008 Substrate proton to heme distances in CYP2C9 allelic variants and alterations by the heterotropic activator, dapsone. Heme 20-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 18485885-4 2008 Co-incubation with dapsone resulted in reduced substrate proton to heme-iron distances for all enzymes, providing at least a partial mechanism for the activation of CYP2C9 variants by dapsone. Dapsone 19-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 18485885-4 2008 Co-incubation with dapsone resulted in reduced substrate proton to heme-iron distances for all enzymes, providing at least a partial mechanism for the activation of CYP2C9 variants by dapsone. Heme 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 18628990-3 2008 Herein, we first evaluated the inhibitory effects of fifteen ginsenosides and sapogenins on human CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4 enzymes by using commercially available fluorescent probes. Ginsenosides 61-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 18485885-4 2008 Co-incubation with dapsone resulted in reduced substrate proton to heme-iron distances for all enzymes, providing at least a partial mechanism for the activation of CYP2C9 variants by dapsone. Iron 72-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 18485885-4 2008 Co-incubation with dapsone resulted in reduced substrate proton to heme-iron distances for all enzymes, providing at least a partial mechanism for the activation of CYP2C9 variants by dapsone. Dapsone 184-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 18485885-5 2008 In summary, neither altered binding affinity nor substrate orientation appear to be major factors in the reduced catalytic activity noted in the CYP2C9 variants, but dapsone co-incubation caused similar changes in substrate proton to heme-iron distances suggesting at least partial common mechanisms in the activation of the CYP2C9 forms. Dapsone 166-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 325-331 18776969-3 2008 Genetic polymorphism of the VKORC1 and CYP2C9 genes, as well as clinical factors such as age, gender, body mass index and interacting drugs explain less than 55% of variability in warfarin dose requirements. Warfarin 180-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 18628990-3 2008 Herein, we first evaluated the inhibitory effects of fifteen ginsenosides and sapogenins on human CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4 enzymes by using commercially available fluorescent probes. Sapogenins 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 18503607-3 2008 Glyburide, a sulphonylurea, is an insulin sensitizer and mainly metabolized by CYP2C9. Glyburide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18503607-3 2008 Glyburide, a sulphonylurea, is an insulin sensitizer and mainly metabolized by CYP2C9. Sulfonylurea Compounds 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18378563-2 2008 Previous in vitro work has demonstrated that genotype-dependent inhibition of CYP2C9 mediated flurbiprofen metabolism, suggesting the possibility of genotype-dependent inhibition interactions in vivo. Flurbiprofen 94-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 18381488-4 2008 Fluconazole is also a moderate inhibitor of CYP2C9 and CYP2C19. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 18385292-8 2008 The P450 probe substrates indicate a decrease in CYP1A2, CYP2B6, CYP2C9, and CYP3A4 activities in HepaRG cells 1 day after removal of DMSO from the medium. Dimethyl Sulfoxide 134-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 18690342-1 2008 The objective of this study was to assess the relative influence of VKORC1 and CYP2C9 genetic variants on several clinical outcomes related to warfarin treatment. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 18690342-7 2008 CYP2C9 alone, VKORC1 alone, and a combination of genetic and clinical factors explained 12%, 27%, and 50%, respectively, of the variation in warfarin maintenance dose. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18183038-0 2008 Prospective study of warfarin dosage requirements based on CYP2C9 and VKORC1 genotypes. Warfarin 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 18183038-1 2008 Polymorphisms in CYP2C9 and VKORC1 have been shown to be associated with warfarin dose requirements and could be used to predict warfarin dose. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 18183038-1 2008 Polymorphisms in CYP2C9 and VKORC1 have been shown to be associated with warfarin dose requirements and could be used to predict warfarin dose. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 18183038-2 2008 We conducted a prospective study in which warfarin dose was prescribed based on CYP2C9 and VKORC1 polymorphisms in 108 Han-Chinese patients without prior warfarin treatments. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 18694831-0 2008 Influence of CYP2C8 and CYP2C9 polymorphisms on pharmacokinetic and pharmacodynamic parameters of racemic and enantiomeric forms of ibuprofen in healthy volunteers. Ibuprofen 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 18576910-7 2008 Warfarin has two enantiomers, R-and S-warfarin, which are substrates primarily of cytochrome P450 (CYP) 3A4 (R-warfarin), CYP1A2 (R-warfarin), and CYP2C9 (S-warfarin). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 18576910-7 2008 Warfarin has two enantiomers, R-and S-warfarin, which are substrates primarily of cytochrome P450 (CYP) 3A4 (R-warfarin), CYP1A2 (R-warfarin), and CYP2C9 (S-warfarin). Ranolazine 30-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 18793590-10 2008 CONCLUSIONS: CYP2C9 activity as measured by apparent oral clearance and urinary metabolic ratio following oral tolbutamide appear similar in people with and without cancer. Tolbutamide 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 18576910-7 2008 Warfarin has two enantiomers, R-and S-warfarin, which are substrates primarily of cytochrome P450 (CYP) 3A4 (R-warfarin), CYP1A2 (R-warfarin), and CYP2C9 (S-warfarin). Sulfur 36-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 18576910-7 2008 Warfarin has two enantiomers, R-and S-warfarin, which are substrates primarily of cytochrome P450 (CYP) 3A4 (R-warfarin), CYP1A2 (R-warfarin), and CYP2C9 (S-warfarin). Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 18694831-7 2008 CYP2C9*3 allele had a decreased racemic ibuprofen metabolism, leading to a 30% augmentation of AUC(0-infinity) and a 30% reduction of clearance compared to CYP2C9*1 (p < 0.05). Ibuprofen 40-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18694831-7 2008 CYP2C9*3 allele had a decreased racemic ibuprofen metabolism, leading to a 30% augmentation of AUC(0-infinity) and a 30% reduction of clearance compared to CYP2C9*1 (p < 0.05). Ibuprofen 40-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 18694831-9 2008 CYP2C9*3 had a 45% reduction of clearance, as well as a 87% and 47% augmentation of AUC(0-infinity) and t(1/2) with respect to CYP2C9*1 of S-ibuprofen and a 30% reduction of clearance of R-ibuprofen. Ibuprofen 139-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18694831-9 2008 CYP2C9*3 had a 45% reduction of clearance, as well as a 87% and 47% augmentation of AUC(0-infinity) and t(1/2) with respect to CYP2C9*1 of S-ibuprofen and a 30% reduction of clearance of R-ibuprofen. Ibuprofen 139-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 18694831-9 2008 CYP2C9*3 had a 45% reduction of clearance, as well as a 87% and 47% augmentation of AUC(0-infinity) and t(1/2) with respect to CYP2C9*1 of S-ibuprofen and a 30% reduction of clearance of R-ibuprofen. Ibuprofen 187-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18694831-9 2008 CYP2C9*3 had a 45% reduction of clearance, as well as a 87% and 47% augmentation of AUC(0-infinity) and t(1/2) with respect to CYP2C9*1 of S-ibuprofen and a 30% reduction of clearance of R-ibuprofen. Ibuprofen 187-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 18694831-12 2008 CONCLUSIONS: This study suggest an impaired metabolism of racemic, S-ibuprofen and R-ibuprofen in CYP2C9*3; an increased R-ibuprofen metabolism in CYP2C8*3; and fewer adverse events in CYP2C8*3 volunteers; that correlates with a decreased expression of iNOS. Ibuprofen 67-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 18694831-12 2008 CONCLUSIONS: This study suggest an impaired metabolism of racemic, S-ibuprofen and R-ibuprofen in CYP2C9*3; an increased R-ibuprofen metabolism in CYP2C8*3; and fewer adverse events in CYP2C8*3 volunteers; that correlates with a decreased expression of iNOS. Ibuprofen 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 18362161-4 2008 In vitro studies have shown that CYP2C19, CYP3A4, and to a lesser extent CYP2C9 contribute to the oxidative metabolism of voriconazole. Voriconazole 122-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 18559094-0 2008 Warfarin dose and INR related to genotypes of CYP2C9 and VKORC1 in patients with myocardial infarction. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 18559094-2 2008 Individual dosage requirement has recently partly been explained by genetic variation of the warfarin metabolizing enzyme CYP2C9 and the Vitamin K-activating enzyme VKORC1. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 18559094-4 2008 AIMS: The primary aim of the present study was to investigate the relation between genotypes of CYP2C9 and VKORC1 and warfarin maintenance dose in myocardial infarction. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 18559094-10 2008 Warfarin dosage requirement was significantly associated with CYP2C9 and VKORC1 genotypes, treatment group and age. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 18559094-12 2008 CONCLUSION: CYP2C9 and VKORC1 genotype frequencies in myocardial infarction patients appear similar to other patient groups and have similar impact on warfarin maintenance dose. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Nelfinavir 129-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Lopinavir 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 18576910-9 2008 The increased warfarin doses required in these two patients may have been caused by induction of CYP3A4 by nevirapine, CYP2C9 by nelfinavir, or CYP2C9 by lopinavir-ritonavir. Ritonavir 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 17851564-3 2008 We hypothesized that capecitabine inhibits the synthesis of CYP2C9, which metabolizes indisulam. Capecitabine 21-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 17851564-3 2008 We hypothesized that capecitabine inhibits the synthesis of CYP2C9, which metabolizes indisulam. N-(3-chloro-7-indolyl)-1,4-benzenedisulphonamide 86-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 18043684-0 2008 The polymorphisms Asn130Asp and Val174Ala in OATP1B1 and the CYP2C9 allele *3 independently affect torsemide pharmacokinetics and pharmacodynamics. Torsemide 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 18496131-10 2008 CONCLUSION: This study indicates that the presently evaluated variant alleles in the CYP2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, GSTA1, GSTP1, ALDH1A1 and ALDH3A1 genes do not explain the interindividual variability in cyclophosphamide and 4-hydroxycyclophosphamide pharmacokinetics and are, probably, not the cause of the observed variability in toxicity. Cyclophosphamide 216-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 18756910-0 2008 Association of CYP2C9 gene polymorphism with bleeding as a complication of warfarin therapy. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 18756910-1 2008 The aim of this study was to determine the association of bleeding as a complication of warfarin therapy with polymorphism of CYP2C9 gene (alleles 1, 2 and 3). Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 18756910-2 2008 The CYP2C9 is the main enzyme for warfarin metabolism. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 18756910-11 2008 Polymorphism of CYP2C9 could determine dose of warfarin therapy and thus it could be related to the risk of bleeding complications. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 18496131-10 2008 CONCLUSION: This study indicates that the presently evaluated variant alleles in the CYP2B6, CYP2C9, CYP2C19, CYP3A4, CYP3A5, GSTA1, GSTP1, ALDH1A1 and ALDH3A1 genes do not explain the interindividual variability in cyclophosphamide and 4-hydroxycyclophosphamide pharmacokinetics and are, probably, not the cause of the observed variability in toxicity. hydroxycyclophosphamide 239-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 18496133-2 2008 CYP2J2, CYP2C8, and CYP2C9 are known to be a source of epoxyeicosatrienoic acids in cardiac tissues. epoxyeicosatrienoic acids 55-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 29783498-0 2008 High carrier prevalence of combinatorial CYP2C9 and VKORC1 genotypes affecting warfarin dosing. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18234403-3 2008 In particularly, molecular analysis of two genes, encoding for the enzyme responsible for the warfarin (S)-isoform catabolism (CYP2C9) and for the target enzyme vitamin K epoxide reductase complex 1 (VKORC1), strongly suggested that their genetic variations greatly affect the individual response to oral anticoagulants. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 18294321-3 2008 CYP2C9 and VKORC1 polymorphisms have been shown to affect warfarin dose requirement. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18305126-3 2008 Tadalafil is mainly metabolized by cytochrome P450 (CYP) 3A4, and as bosentan induces CYP2C9 and CYP3A4, a pharmacokinetic interaction is possible between these agents. Bosentan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 18356043-2 2008 Screening with well-established chemical inhibitors showed that the formation of norethindrone was potently inhibited by CYP3A4 inhibitor ketoconazole (IC(50)=0.02 microM) and with CYP2C9 inhibitor sulphaphenazole (IC(50)=2.13 microM); the further biotransformation of norethindrone was strongly inhibited by ketoconazole (IC(50)=0.09 microM). Norethindrone 81-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 18356043-2 2008 Screening with well-established chemical inhibitors showed that the formation of norethindrone was potently inhibited by CYP3A4 inhibitor ketoconazole (IC(50)=0.02 microM) and with CYP2C9 inhibitor sulphaphenazole (IC(50)=2.13 microM); the further biotransformation of norethindrone was strongly inhibited by ketoconazole (IC(50)=0.09 microM). Sulfaphenazole 198-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 18356043-4 2008 Lynestrenol bioactivation was mainly catalysed by recombinant human CYP2C9, CYP2C19 and CYP3A4; rCYP3A4 was responsible for the hydroxylation of norethindrone. Lynestrenol 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 18356043-4 2008 Lynestrenol bioactivation was mainly catalysed by recombinant human CYP2C9, CYP2C19 and CYP3A4; rCYP3A4 was responsible for the hydroxylation of norethindrone. Norethindrone 145-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 18356043-5 2008 A significant correlation was observed between norethindrone formation and tolbutamide hydroxylation, a CYP2C9-selective activity (r=0.63; p=0.01). Norethindrone 47-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 18356043-5 2008 A significant correlation was observed between norethindrone formation and tolbutamide hydroxylation, a CYP2C9-selective activity (r=0.63; p=0.01). Tolbutamide 75-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 18570163-0 2008 Role of pharmacodiagnostic of CYP2C9 variants in the optimization of warfarin therapy in Malaysia: a 6-month follow-up study. Warfarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 18570163-2 2008 A retrospective study was conducted to explore the importance of CYP2C9 genotyping for the initiation and maintenance therapy of warfarin in clinical practice. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 18030307-0 2008 Combination of phenotype assessments and CYP2C9-VKORC1 polymorphisms in the determination of warfarin dose requirements in heavily medicated patients. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 18030307-8 2008 Thus, combination of a phenotypic measure to CYP2C9-VKORC1 genotypes represents a useful strategy to predict warfarin doses in patients receiving multiple drugs (11+/-4 drugs/day). Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 18256202-0 2008 The "albumin effect" and in vitro-in vivo extrapolation: sequestration of long-chain unsaturated fatty acids enhances phenytoin hydroxylation by human liver microsomal and recombinant cytochrome P450 2C9. long-chain unsaturated fatty acids 74-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-203 18256202-0 2008 The "albumin effect" and in vitro-in vivo extrapolation: sequestration of long-chain unsaturated fatty acids enhances phenytoin hydroxylation by human liver microsomal and recombinant cytochrome P450 2C9. Phenytoin 118-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-203 18256202-2 2008 BSA and essentially fatty acid-free human serum albumin (HSA-FAF) reduced the K(m) values for PHY hydroxylation (based on unbound substrate concentration) by human liver microsomes (HLMs) and recombinant CYP2C9 by approximately 75%, with only a minor effect on V(max). Fatty Acids 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 204-210 18256202-5 2008 A mixture of arachidonic, linoleic and oleic acids, at a concentration corresponding to 1/20 of the content of HLMs, doubled the K(m) for PHY hydroxylation by CYP2C9, without affecting V(max). arachidonic 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 18256202-5 2008 A mixture of arachidonic, linoleic and oleic acids, at a concentration corresponding to 1/20 of the content of HLMs, doubled the K(m) for PHY hydroxylation by CYP2C9, without affecting V(max). Linoleic Acid 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 18256202-5 2008 A mixture of arachidonic, linoleic and oleic acids, at a concentration corresponding to 1/20 of the content of HLMs, doubled the K(m) for PHY hydroxylation by CYP2C9, without affecting V(max). Oleic Acids 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 18256202-7 2008 K(i) values for arachidonic acid inhibition of human liver microsomal- and CYP2C9-catalyzed PHY hydroxylation were 3.8 and 1.6 microM, respectively. Arachidonic Acid 16-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 18356043-9 2008 Both lynestrenol and norethindrone were weak inhibitors of CYP2C9 (IC(50) of 32 microM and 46 microM for tolbutamide hydroxylation, respectively). Lynestrenol 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 18356043-9 2008 Both lynestrenol and norethindrone were weak inhibitors of CYP2C9 (IC(50) of 32 microM and 46 microM for tolbutamide hydroxylation, respectively). Norethindrone 21-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 18356043-9 2008 Both lynestrenol and norethindrone were weak inhibitors of CYP2C9 (IC(50) of 32 microM and 46 microM for tolbutamide hydroxylation, respectively). Tolbutamide 105-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 18356043-10 2008 In conclusion, CYP2C9, CYP2C19 and CYP3A4 are the primary cytochromes in the bioactivation of lynestrenol in vitro, while CYP3A4 catalyses the further metabolism of norethindrone. Lynestrenol 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 18356043-10 2008 In conclusion, CYP2C9, CYP2C19 and CYP3A4 are the primary cytochromes in the bioactivation of lynestrenol in vitro, while CYP3A4 catalyses the further metabolism of norethindrone. Norethindrone 165-178 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 18466099-0 2008 Influence of CYP2C9 and VKORC1 on warfarin dose, anticoagulation attainment and maintenance among European-Americans and African-Americans. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 18466099-4 2008 RESULTS & CONCLUSION: CYP2C9 and VKORC1 accounted for up to 30% of the variability in warfarin dose among European-Americans and 10% among African-Americans. Adenosine Monophosphate 9-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 18466099-4 2008 RESULTS & CONCLUSION: CYP2C9 and VKORC1 accounted for up to 30% of the variability in warfarin dose among European-Americans and 10% among African-Americans. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 29783498-1 2008 BACKGROUND: Polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) genes significantly alter the effective warfarin dose. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-52 29783498-1 2008 BACKGROUND: Polymorphisms in the cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) genes significantly alter the effective warfarin dose. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 29783498-2 2008 The CYP2C9*2 (430C>T), CYP2C9*3 (1075A>C) and VKORC1 -1639 G>A polymorphisms affect warfarin dose through altered metabolism (CYP2C9) and sensitivity (VKORC1). Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 29783498-2 2008 The CYP2C9*2 (430C>T), CYP2C9*3 (1075A>C) and VKORC1 -1639 G>A polymorphisms affect warfarin dose through altered metabolism (CYP2C9) and sensitivity (VKORC1). Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 29783498-2 2008 The CYP2C9*2 (430C>T), CYP2C9*3 (1075A>C) and VKORC1 -1639 G>A polymorphisms affect warfarin dose through altered metabolism (CYP2C9) and sensitivity (VKORC1). Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 29783498-9 2008 Combinatorial genotyping of CYP2C9 and VKORC1 can allow for individualized dosing of warfarin amongst patients with gene polymorphisms potentially reducing the risk of accentuated responses and bleeding. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 18204476-0 2008 Effects of St John"s wort and CYP2C9 genotype on the pharmacokinetics and pharmacodynamics of gliclazide. Gliclazide 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 18634396-8 2008 A common mutation in the gene coding for the cytochrome P450 (CYP2C9), with one or more combinations of its polymorphisms, is responsible for the reduced warfarin requirements or for the resistance to warfarin. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 18634396-8 2008 A common mutation in the gene coding for the cytochrome P450 (CYP2C9), with one or more combinations of its polymorphisms, is responsible for the reduced warfarin requirements or for the resistance to warfarin. Warfarin 201-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 18322281-1 2008 BACKGROUND: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9), and of a key pharmacologic target of warfarin, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses, but the role of these variants during initial anticoagulation is not clear. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-90 17868191-2 2008 Carriers of CYP2C9 mutant alleles exhibit a diminished CYP2C9 metabolic capacity leading to decreased endothelium-derived hyperpolarizing factors (EDHF) synthesis and an increased risk for atherosclerosis. hyperpolarizing factors 122-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17868191-2 2008 Carriers of CYP2C9 mutant alleles exhibit a diminished CYP2C9 metabolic capacity leading to decreased endothelium-derived hyperpolarizing factors (EDHF) synthesis and an increased risk for atherosclerosis. hyperpolarizing factors 122-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 17868191-2 2008 Carriers of CYP2C9 mutant alleles exhibit a diminished CYP2C9 metabolic capacity leading to decreased endothelium-derived hyperpolarizing factors (EDHF) synthesis and an increased risk for atherosclerosis. edhf 147-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17868191-2 2008 Carriers of CYP2C9 mutant alleles exhibit a diminished CYP2C9 metabolic capacity leading to decreased endothelium-derived hyperpolarizing factors (EDHF) synthesis and an increased risk for atherosclerosis. edhf 147-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 18702389-0 2008 Acenocoumarol and phenytoin toxicity in the presence of CYP2C9 mutation. Acenocoumarol 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 18702389-0 2008 Acenocoumarol and phenytoin toxicity in the presence of CYP2C9 mutation. Phenytoin 18-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 18315785-1 2008 BACKGROUND AND OBJECTIVE: CYP2C9 is a polymorphic enzyme that has been reported to metabolize several clinically useful drugs such as warfarin, phenytoin and non-steroidal anti-inflammatory drugs. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 18315785-1 2008 BACKGROUND AND OBJECTIVE: CYP2C9 is a polymorphic enzyme that has been reported to metabolize several clinically useful drugs such as warfarin, phenytoin and non-steroidal anti-inflammatory drugs. Phenytoin 144-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 18367977-9 2008 Of all the AEDs, only phenytoin undergoes significant metabolism by cytochrome P450 isozymes with significant genetic polymorphisms (CYP2C9, CYP2C19). Phenytoin 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 18311908-4 2008 In this study, the structural characteristics of nitrogen-containing compounds, which bind to the iron atom in two CYP isoforms (CYP2C9 and CYP3A4), were investigated. Nitrogen 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 18311908-4 2008 In this study, the structural characteristics of nitrogen-containing compounds, which bind to the iron atom in two CYP isoforms (CYP2C9 and CYP3A4), were investigated. Iron 98-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 18206980-5 2008 Among the several CYPs tested, CYP2C9/2C19 and 1A2 were the most efficient in EPA and DHA epoxidations. Eicosapentaenoic Acid 78-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18206980-5 2008 Among the several CYPs tested, CYP2C9/2C19 and 1A2 were the most efficient in EPA and DHA epoxidations. Docosahexaenoic Acids 86-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 18206980-6 2008 It ensued that 10muM of these two omega3 fatty acids decreased by more than 80% and 60%, respectively, the formation by CYP2C9 of AA-epoxidised derivatives. Fatty Acids, Omega-3 34-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 18322281-7 2008 CONCLUSIONS: Initial variability in the INR response to warfarin was more strongly associated with genetic variability in the pharmacologic target of warfarin, VKORC1, than with CYP2C9. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 18322281-1 2008 BACKGROUND: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9), and of a key pharmacologic target of warfarin, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses, but the role of these variants during initial anticoagulation is not clear. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 18322281-1 2008 BACKGROUND: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9), and of a key pharmacologic target of warfarin, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses, but the role of these variants during initial anticoagulation is not clear. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-90 18322281-1 2008 BACKGROUND: Genetic variants of the enzyme that metabolizes warfarin, cytochrome P-450 2C9 (CYP2C9), and of a key pharmacologic target of warfarin, vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses, but the role of these variants during initial anticoagulation is not clear. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 18322281-6 2008 Both the CYP2C9 genotype and VKORC1 haplotype had a significant influence on the required warfarin dose after the first 2 weeks of therapy. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 17226852-0 2008 Oral anticoagulation with warfarin is significantly influenced by steroids and CYP2C9 polymorphisms in children with cancer. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 17851566-0 2008 CYP2C9 genotype-guided warfarin prescribing enhances the efficacy and safety of anticoagulation: a prospective randomized controlled study. Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17851566-1 2008 Warfarin anticoagulation effect is characterized by marked variability, some of which has been attributed to CYP2C9 polymorphisms. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 17851566-2 2008 This study prospectively examines whether a priori knowledge of CYP2C9 genotype may improve warfarin therapy. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 17851566-7 2008 In conclusion, CYP2C9 genotype-guided warfarin therapy is more efficient and safer than the "average-dose" protocol. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 17999057-8 2008 All are eliminated by hepatic metabolism involving mainly CYP2C9 (celecoxib, lumiracoxib) and CYP3A4 (etoricoxib). Celecoxib 66-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 17999057-8 2008 All are eliminated by hepatic metabolism involving mainly CYP2C9 (celecoxib, lumiracoxib) and CYP3A4 (etoricoxib). lumiracoxib 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 17226852-5 2008 Thus, awareness of CYP2C9 genotype and steroid-induced responsiveness to warfarin may be important when administrating oral anticoagulation in children. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 29783347-6 2008 Pharmacogenetic studies have determined that variants in the CYP2C9 and VKORC1 genes help to predict the therapeutic warfarin dose. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 18303964-1 2008 UNLABELLED: The cytochrome P450 enzymes CYP2C8, CYP2C9 and CYP2J2 generate 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid (EET) from arachidonic acid, and these EETs are then hydrolyzed to dihydroxyeicosatrienoic acids (DHET) before excretion into the urine. 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid 75-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 18303964-1 2008 UNLABELLED: The cytochrome P450 enzymes CYP2C8, CYP2C9 and CYP2J2 generate 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid (EET) from arachidonic acid, and these EETs are then hydrolyzed to dihydroxyeicosatrienoic acids (DHET) before excretion into the urine. EET 124-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 18303964-1 2008 UNLABELLED: The cytochrome P450 enzymes CYP2C8, CYP2C9 and CYP2J2 generate 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid (EET) from arachidonic acid, and these EETs are then hydrolyzed to dihydroxyeicosatrienoic acids (DHET) before excretion into the urine. Arachidonic Acid 134-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 18303964-1 2008 UNLABELLED: The cytochrome P450 enzymes CYP2C8, CYP2C9 and CYP2J2 generate 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid (EET) from arachidonic acid, and these EETs are then hydrolyzed to dihydroxyeicosatrienoic acids (DHET) before excretion into the urine. dihydroxyeicosatrienoic acids 190-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 18252229-0 2008 Warfarin pharmacogenetics: CYP2C9 and VKORC1 genotypes predict different sensitivity and resistance frequencies in the Ashkenazi and Sephardi Jewish populations. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 18303964-1 2008 UNLABELLED: The cytochrome P450 enzymes CYP2C8, CYP2C9 and CYP2J2 generate 8,9-, 11,12-, and 14,15-epoxyeicosatrienic acid (EET) from arachidonic acid, and these EETs are then hydrolyzed to dihydroxyeicosatrienoic acids (DHET) before excretion into the urine. dhet 221-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 18274956-6 2008 Usnic acid was a relatively weak inhibitor of CYP2D6 and a potent inhibitor of CYP2C19 (the concentration eliciting 50% inhibition (IC(50)) = 9 nM) and CYP2C9 (IC(50) = 94 nM), with less potent inhibition of CYP2C8 (IC(50) = 1.9 microM) and CYP2C18 (IC(50) = 6.3 microM). usnic acid 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 18443385-3 2008 Hence, there are chances of drug-drug interaction because modulations of isoenzymes involved in metabolism CYP2C9/10 and CYP2C19 which partially inhibited by etoricoxib. Etoricoxib 158-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 18237107-2 2008 The molecular alignment program ROCS was used with the query molecule flurbiprofen as a basis for predicting the correct active site orientation of the CYP2C9 database molecules. Flurbiprofen 70-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 18237107-4 2008 Transposition of the first 39 correctly aligned molecules into the CYP2C9 active site yielded an average site of metabolism to iron heme distance of 5.21 A, in good agreement with previous experimental observations. iron heme 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 18445991-0 2008 The effect of bucolome, a CYP2C9 inhibitor, on the pharmacokinetics of losartan. Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 17597710-1 2008 Sulfonylurea hypoglycemics are mainly metabolized by the cytochrome P450 2C9 (CYP2C9) enzyme. Sulfonylurea Compounds 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-76 17597710-1 2008 Sulfonylurea hypoglycemics are mainly metabolized by the cytochrome P450 2C9 (CYP2C9) enzyme. Sulfonylurea Compounds 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 17597710-4 2008 Associations between CYP2C9 polymorphisms, prescribed doses of sulfonylurea, and change in glucose levels after the start of sulfonylurea therapy were assessed in all patients with incident diabetes mellitus starting on sulfonylurea therapy in the Rotterdam Study, a population-based cohort study of 7,983 elderly people. Glucose 91-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 17597710-5 2008 In CYP2C9*3 allele carriers using tolbutamide, the prescribed dose was lower compared to patients with the wild-type CYP2C9 genotype. Tolbutamide 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 17597710-5 2008 In CYP2C9*3 allele carriers using tolbutamide, the prescribed dose was lower compared to patients with the wild-type CYP2C9 genotype. Tolbutamide 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 17597710-7 2008 In CYP2C9*3 allele carriers, the mean decrease in fasting serum glucose levels after the start of tolbutamide therapy was larger than in patients with the wild-type genotype, although not statistically significant. Glucose 64-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 17597710-7 2008 In CYP2C9*3 allele carriers, the mean decrease in fasting serum glucose levels after the start of tolbutamide therapy was larger than in patients with the wild-type genotype, although not statistically significant. Tolbutamide 98-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 17597710-8 2008 Patients with diabetes mellitus who are carriers of a CYP2C9*3 allele require lower doses of tolbutamide to regulate their serum glucose levels compared to patients with the wild-type genotype. Tolbutamide 93-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17597710-8 2008 Patients with diabetes mellitus who are carriers of a CYP2C9*3 allele require lower doses of tolbutamide to regulate their serum glucose levels compared to patients with the wild-type genotype. Glucose 129-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 18281922-0 2008 Pharmacogenetic testing of CYP2C9 and VKORC1 alleles for warfarin. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 18281922-8 2008 Variants of two genes, CYP2C9 and VKORC1, account for 30-50% of the variability in dosing of warfarin; thus, many believe that testing of these genes will aid in warfarin dosing recommendations. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 18281922-8 2008 Variants of two genes, CYP2C9 and VKORC1, account for 30-50% of the variability in dosing of warfarin; thus, many believe that testing of these genes will aid in warfarin dosing recommendations. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 18281922-10 2008 In an effort to address this situation, a multidisciplinary expert group was organized in November 2006 to evaluate the role of CYP2C9 and VKORC1 testing in altering warfarin-related therapeutic goals and reduction of adverse drug events. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 18281922-16 2008 Although the routine use of warfarin genotyping is not endorsed by this work group at this time, in certain situations, CYP2C9 and VKORC1 testing may be useful, and warranted, in determining the cause of unusual therapeutic responses to warfarin therapy. Warfarin 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 18370846-0 2008 Dosing algorithm for warfarin using CYP2C9 and VKORC1 genotyping from a multi-ethnic population: comparison with other equations. Warfarin 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 18370846-1 2008 OBJECTIVES: Polymorphism in the genes for cytochrome (CYP)2C9 and the vitamin K epoxide reductase complex subunit 1 (VKORC1) affect the pharmacokinetics and pharmacodynamics of warfarin. Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-61 18370846-3 2008 METHODS: We determined the allele and haplotype frequencies of genes for CYP2C9 and VKORC1 on 167 Caucasian, African-American, Asian and Hispanic patients on warfarin. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 19099951-0 2008 [Association between CYP2C9 and VKORC1 genetic polymorphism and warfarin dose requirements]. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 19099951-8 2008 The multiple linear regression model for warfarin dose indicated age, weight and VKORC1 genotype could explain the inter-individual variation in dose requirement of 9.3%, 7.4%, 51.9% patients, respectively; age, weight, CYP2C9 and VKORC1 genotype together could explain the inter-individual variation in dose requirement of 64.1% patients. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 19099951-9 2008 CONCLUSION: This study showed that age, weight and VKORC1 and CYP2C9 polymorphism had significant influences on warfarin dose requirements and should be considered on dosing regimens modification to improve the safety of warfarin therapy. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 19099951-9 2008 CONCLUSION: This study showed that age, weight and VKORC1 and CYP2C9 polymorphism had significant influences on warfarin dose requirements and should be considered on dosing regimens modification to improve the safety of warfarin therapy. Warfarin 221-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 17975109-7 2008 EET production by human recombinant CYP 2C9 and 2J2, 2 major epoxygenase isozymes expressed in human coronary arterioles, was directly inhibited in a concentration-dependent fashion by H2O2 in vitro, as observed by high-performance liquid chromatography (HPLC); however, EETs were not directly sensitive to oxidative modification. Hydrogen Peroxide 185-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-43 17895500-0 2008 Variant CYP2C9 alleles and warfarin concentrations in patients receiving low-dose versus average-dose warfarin therapy. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 17895500-7 2008 A warfarin requirement of <2.5 mg/day and an elevated warfarin S/R concentration ratio were each associated with a higher frequency of variant CYP2C9 alleles. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 17895500-7 2008 A warfarin requirement of <2.5 mg/day and an elevated warfarin S/R concentration ratio were each associated with a higher frequency of variant CYP2C9 alleles. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 18698879-8 2008 Compared with homozygous carriers of CYP2C9*1, patients homozygous for CYP2C9*3 were estimated to need 3.3-fold lower mean doses of warfarin to achieve the same international normalized ratio, with *2 carriers and heterozygous patients in between. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 18698879-8 2008 Compared with homozygous carriers of CYP2C9*1, patients homozygous for CYP2C9*3 were estimated to need 3.3-fold lower mean doses of warfarin to achieve the same international normalized ratio, with *2 carriers and heterozygous patients in between. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 18788834-13 2008 Any greater than expected increases in bleeding secondary to anticoagulation may potentially offset any benefit gained from cardioembolic stroke reduction, although this has not been fully resolved.Finally, there are racial/ethnic differences in the prevalence of certain polymorphisms in genes that influence warfarin pharmacokinetics and pharmacodynamics (e.g. cytochrome P450 2C9 and vitamin K epoxide reductase). Warfarin 310-318 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 363-382 19026034-4 2008 Systemic triazoles are inhibitors of cytochrome P450 (CYP) isozymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Triazoles 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 18636784-8 2008 The half-life of benzbromarone is generally short (about 3 hours); however, a uricosuric metabolite, 6-hydroxybenzbromarone, has a much longer half-life (up to 30 hours) and is the major species responsible for the uricosuric activity of benzbromarone, although its metabolism by cytochrome P450 (CYP) 2C9 in the liver may vary between patients as a result of polymorphisms in this enzyme. 6-hydroxybenzbromarone 101-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 280-305 17687273-6 2008 The most likely mechanism for the interaction between terbinafine and venlafaxine is the inhibition of CYP2D6-mediated O-demethylation of venlafaxine, whereas the minor effects of voriconazole are probably due to the inhibition of CYP3A4-, CYP2C9-, or CYP2C19-mediated metabolism of venlafaxine. Terbinafine 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 17687273-6 2008 The most likely mechanism for the interaction between terbinafine and venlafaxine is the inhibition of CYP2D6-mediated O-demethylation of venlafaxine, whereas the minor effects of voriconazole are probably due to the inhibition of CYP3A4-, CYP2C9-, or CYP2C19-mediated metabolism of venlafaxine. Venlafaxine Hydrochloride 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 17906972-2 2008 SNPs in the cytochrome P450 complex (CYP2C9) affect warfarin metabolism: patients who have the CYP2C9*2 and/or CYP2C9*3 variants metabolize warfarin slowly and are more likely to have an elevated International Normalized Ratio INR or to hemorrhage during warfarin initiation than patients without these variants. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 18399713-12 2008 CONCLUSION: This study shows that the CYP2C9*3 and SLCO1B1 c.521TC genotype and female gender are significant and independent predictors of the pharmacokinetics of torasemide. Torsemide 164-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. Carbamazepine 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. Carbamazepine 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. Sulfamethoxazole 210-226 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. Sulfamethoxazole 210-226 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. prohapten 230-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 18991696-7 2008 Carbamazepine hypersensitivity is also influenced by gene variants of cytochrome P450 enzymes on the generation of reactive metabolites, while CYP2C9*2 and CYP2C9*3 polymorphisms influence the bioactivation of sulfamethoxazole in prohapten. prohapten 230-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 18445991-1 2008 Losartan, a selective angiotensin receptor antagonist, is mainly metabolized by CYP2C9 to an active carboxylic acid, E3174, which is pharmacologically more potent inhibitor than the parent compound. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 18445991-1 2008 Losartan, a selective angiotensin receptor antagonist, is mainly metabolized by CYP2C9 to an active carboxylic acid, E3174, which is pharmacologically more potent inhibitor than the parent compound. Carboxylic Acids 100-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 18445991-1 2008 Losartan, a selective angiotensin receptor antagonist, is mainly metabolized by CYP2C9 to an active carboxylic acid, E3174, which is pharmacologically more potent inhibitor than the parent compound. losartan carboxylic acid 117-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 18445991-2 2008 We evaluated the effect of bucolome, a CYP2C9 inhibitor, on the pharmacokinetics of losartan and E3174, which were measured by high performance liquid chromatography in human volunteers and rats. bucolome 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 18636784-8 2008 The half-life of benzbromarone is generally short (about 3 hours); however, a uricosuric metabolite, 6-hydroxybenzbromarone, has a much longer half-life (up to 30 hours) and is the major species responsible for the uricosuric activity of benzbromarone, although its metabolism by cytochrome P450 (CYP) 2C9 in the liver may vary between patients as a result of polymorphisms in this enzyme. Benzbromarone 110-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 280-305 18636784-15 2008 If all the reported cases are assumed to be due to benzbromarone, the estimated risk of hepatotoxicity in Europe was approximately 1 in 17 000 patients but may be higher in Japan.Benzbromarone is also an inhibitor of CYP2C9 and so may be involved in drug interactions with drugs dependent on this enzyme for clearance, such as warfarin. Benzbromarone 51-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 18636784-15 2008 If all the reported cases are assumed to be due to benzbromarone, the estimated risk of hepatotoxicity in Europe was approximately 1 in 17 000 patients but may be higher in Japan.Benzbromarone is also an inhibitor of CYP2C9 and so may be involved in drug interactions with drugs dependent on this enzyme for clearance, such as warfarin. Benzbromarone 179-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 18636784-15 2008 If all the reported cases are assumed to be due to benzbromarone, the estimated risk of hepatotoxicity in Europe was approximately 1 in 17 000 patients but may be higher in Japan.Benzbromarone is also an inhibitor of CYP2C9 and so may be involved in drug interactions with drugs dependent on this enzyme for clearance, such as warfarin. Warfarin 327-335 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 19074093-1 2008 Polymorphisms in CYP2C9, a critical cytochrome P-450 enzyme in the metabolism of warfarin, alters its clearance and affects dosing. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 18429757-0 2008 [Cytochrome P4502C9(CYP2C9) gene polymorphism and safety of therapy with warfarin]. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 17955230-1 2008 OBJECTIVE: To evaluate the impact of the two most common CYP2C9 variant alleles (*2 and *3) on the maintenance dose of warfarin and on the quality of anticoagulation control in Brazilians. Warfarin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 17955230-9 2008 Patients with CYP2C9*3, but not CYP2C9*2, required significantly (one-sided P = 0.001, Mann-Whitney U-test) lower warfarin maintenance doses (3.1 +/- 1.8 mg) than CYP2C9*1*1 patients (5.3 +/- 2.1 mg). Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 17955230-10 2008 CONCLUSION: Patients with either CYP2C9*2 or CYP2C9*3 show higher risk of over-anticoagulation compared to CYP2C9*1*1 subjects and could benefit from a reduction in the initial warfarin standard dose (e.g., to 2.5 mg/day). Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 17955230-10 2008 CONCLUSION: Patients with either CYP2C9*2 or CYP2C9*3 show higher risk of over-anticoagulation compared to CYP2C9*1*1 subjects and could benefit from a reduction in the initial warfarin standard dose (e.g., to 2.5 mg/day). Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 17955230-10 2008 CONCLUSION: Patients with either CYP2C9*2 or CYP2C9*3 show higher risk of over-anticoagulation compared to CYP2C9*1*1 subjects and could benefit from a reduction in the initial warfarin standard dose (e.g., to 2.5 mg/day). Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 18025525-7 2008 The decrease in voriconazole exposure during coadministration is probably mainly due to the induction of CYP2C19 and CYP2C9 by efavirenz. Voriconazole 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 18429757-1 2008 Aim of the study was to investigate frequency and influence of alleles CYP2C9*2 and CYP2C9*3 on pharmacokinetics, pharmacodynamics and dosing regimen of warfarin and on development of hemorrhagic complications. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 18836275-1 2008 OBJECTIVE: The aim of the present study was to investigate the role of CYP2C9 gene polymorphisms after heart valve replacement in a group of patients on warfarin therapy. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 18836275-5 2008 RESULTS: The patients with CYP2C9*1/*3 and CYP2C9*2/*3 genotypes were taking 28.21 and 24.47 mg, respectively, as mean weekly warfarin dose, whereas patients with CYP2C9*1/*1 genotype were taking 33.90 mg. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 18429757-1 2008 Aim of the study was to investigate frequency and influence of alleles CYP2C9*2 and CYP2C9*3 on pharmacokinetics, pharmacodynamics and dosing regimen of warfarin and on development of hemorrhagic complications. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 18836275-5 2008 RESULTS: The patients with CYP2C9*1/*3 and CYP2C9*2/*3 genotypes were taking 28.21 and 24.47 mg, respectively, as mean weekly warfarin dose, whereas patients with CYP2C9*1/*1 genotype were taking 33.90 mg. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 18836275-6 2008 CONCLUSION: The data show that patients with CYP2C9*1/*3 and CYP2C9*2/*3 genotypes needed a lower maintenance dose of warfarin than patients with CYP2C9*1/*1 wild-type genotype. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 18836275-6 2008 CONCLUSION: The data show that patients with CYP2C9*1/*3 and CYP2C9*2/*3 genotypes needed a lower maintenance dose of warfarin than patients with CYP2C9*1/*1 wild-type genotype. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 18429757-6 2008 Average maintenance doses of warfarin for patients with allele variants CYP 2C9 *2 and 2C9 *3 were 3.6 and 3.1 mg/day, respectively, what was significantly lower than in wild type homozygotes (6.1 mg/day). Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-79 18836275-6 2008 CONCLUSION: The data show that patients with CYP2C9*1/*3 and CYP2C9*2/*3 genotypes needed a lower maintenance dose of warfarin than patients with CYP2C9*1/*1 wild-type genotype. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 17899045-1 2007 OBJECTIVE: The objective of this study was to assess the contribution of the VKORC1 and CYP2C9 genotypes and age, body size, and weight of the patients to the warfarin dose requirement in a Chinese population. Warfarin 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 18374198-5 2008 SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of warfarin. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-27 18374198-5 2008 SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of warfarin. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 18374198-6 2008 The combined analysis of VKORC1, CYP2C9 SNPs, and age may account for more than 50% of the individual variability in the warfarin maintenance dosage. Warfarin 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 18159128-1 2007 Nateglinide is an antidiabetic agent metabolized by CYP2C9 and CYP3A4; hence inhibitors of these CYP isozymes may interact with nateglinide. Nateglinide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 18159128-1 2007 Nateglinide is an antidiabetic agent metabolized by CYP2C9 and CYP3A4; hence inhibitors of these CYP isozymes may interact with nateglinide. Nateglinide 128-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 17951033-4 2007 However, in microsomes prepared from native human hepatocytes, only relatively specific competitors (inhibitors and/or substrates) of CYP1A2, CYP2C8, CYP2C9 and CYP3A4 reduced notably the degradation cyamemazine. cyamemazine 200-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 17951033-5 2007 The main routes of cyamemazine biotransformation are N-mono-demethylation (CYP1A2, CYP3A4 and CYP2C8) and mono-oxidation (either S-oxidized or hydroxylated derivatives which could not be discriminated because characterized by the same mass value) by CYP1A2 and CYP2C9. cyamemazine 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 261-267 17872452-3 2007 METHODS AND RESULTS: In native CYP2C-expressing endothelial cells, bradykinin elicited a Ca(2+) influx that was potentiated by the soluble epoxide hydrolase inhibitor, 1-adamantyl-3-cyclohexylurea (ACU), and attenuated by CYP inhibition. CHEMBL242255 168-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-36 17872452-4 2007 Similar effects were observed in cultured endothelial cells overexpressing CYP2C9, but not in CYP2C9-deficient cells, and were prevented by the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid as well as by the cAMP antagonist, Rp-cAMPS. EET 144-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 17872452-4 2007 Similar effects were observed in cultured endothelial cells overexpressing CYP2C9, but not in CYP2C9-deficient cells, and were prevented by the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid as well as by the cAMP antagonist, Rp-cAMPS. 14,15-episulfide eicosatrienoic acid 159-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 17872452-4 2007 Similar effects were observed in cultured endothelial cells overexpressing CYP2C9, but not in CYP2C9-deficient cells, and were prevented by the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid as well as by the cAMP antagonist, Rp-cAMPS. Cyclic AMP 211-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 18020424-5 2007 CYP2C9 was the major recombinant enzyme capable of catalyzing the formation of 6-OH BBR, although CYP2C19 also showed a lower degree of activity. 6-oh bbr 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 18020424-9 2007 Isoform profiling with recombinant human P450s suggested that CYP2C9 is primarily responsible for the formation of this reactive quinone intermediate. quinone 129-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 17899045-7 2007 The multiple linear regression model for warfarin dose indicated significant contributions from age (r (2) = 0.084; P < 0.001), weight (r (2) = 0.063; P < 0.001), VKORC1 genotype (r (2) = 0.494; P < 0.001), and age, weight, and CYP2C9 and VKORC1 genotype together (r (2) = 0.628; P < 0.001). Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 237-243 17899045-8 2007 CONCLUSION: This study shows that age, weight and the VKORC1 and CYP2C9 polymorphism affect warfarin dose requirements in our sample of Chinese patients receiving long-term therapy and showing stable control of anticoagulation. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17899045-9 2007 It is anticipated that the use of dosing regimens modified by taking into account the contribution of age, weight, and the CYP2C9 and VKORC1 genotypes has the potential to improve the safety of warfarin therapy. Warfarin 194-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 18034666-1 2007 The nonsteroidal anti-inflammatory drug diclofenac is extensively metabolized by cytochrome P450 (CYP) enzymes, mainly by CYP2C9. Diclofenac 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 17904094-9 2007 Turnover rates for luciferin ME and H metabolism by CYP2C9 and mutants were at least one order of magnitude higher in experiments with membranes compared to whole cells, consistent with impaired product egress from cells. D-luciferin 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 18034666-11 2007 The inhibition of CYP2C9, and to some extent that of CYP3A4 and CYP2C19 enzymes during the first-pass metabolism of diclofenac seems to be involved in the interaction. Diclofenac 116-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 18021343-0 2007 Pharmacogenetics of acenocoumarol: CYP2C9, CYP2C19, CYP1A2, CYP3A4, CYP3A5 and ABCB1 gene polymorphisms and dose requirements. Acenocoumarol 20-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 17900275-0 2007 Common polymorphisms of CYP2C19 and CYP2C9 affect the pharmacokinetic and pharmacodynamic response to clopidogrel but not prasugrel. Clopidogrel 102-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 17900275-8 2007 CONCLUSIONS: The common loss of function polymorphisms of CYP2C19 and CYP2C9 are associated with decreased exposure to the active metabolite of clopidogrel but not prasugrel. Clopidogrel 144-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 17900275-8 2007 CONCLUSIONS: The common loss of function polymorphisms of CYP2C19 and CYP2C9 are associated with decreased exposure to the active metabolite of clopidogrel but not prasugrel. Prasugrel Hydrochloride 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 17904094-11 2007 Finally screening with a panel of CYP2C ligands using CYP2C9 or active mutants revealed different patterns of inhibition and heteroactivation of metabolism of luciferin analogs. D-luciferin 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 18034618-2 2007 Our primary objective was to determine whether the genes for cytochrome P450 (CYP) 2C9, nicotinamide adenine dinucleotide phosphate, reduced, quinone oxidoreductase (NQO1) and vitamin K epoxide reductase complex subunit 1 (VKORC1) are associated with warfarin dose requirements in African-Americans. Warfarin 251-259 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-86 18034619-7 2007 Also, CYP2C9*2 (Arg144Cys) and CYP2C9*3 (Ile359Leu) variant alleles were significantly associated with low MWWD (p = 0.003 and 0.027, respectively). ile359leu 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 70-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 70-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 70-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 174-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 174-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 17686967-1 2007 CYP2C9 catalyzes the demethylation of the biphasic kinetics substrate (S)-naproxen, and the CYP2C9*2 (R144C) and CYP2C9*3 (I359L) variants are associated with lower rates of (S)-naproxen demethylation. Naproxen 174-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 17686967-4 2007 Uncoupling in CYP2C9.1 and CYP2C9.3 was primarily to H(2)O(2). Hydrogen Peroxide 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 17686967-4 2007 Uncoupling in CYP2C9.1 and CYP2C9.3 was primarily to H(2)O(2). Hydrogen Peroxide 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 17686967-5 2007 In contrast, CYP2C9.2 uncoupled to excess water preferentially. Water 42-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 18034618-5 2007 Possession of a CYP2C9*2, *3 or *5 allele was associated with a 38% lower warfarin dose compared with the *1/*1 genotype (30 +/- 13 vs 48 +/- 18 mg/week; p = 0.003). Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 18034618-7 2007 Multiple regression analysis revealed that CYP2C9 genotype (p = 0.015), age (p < 0.001) and body surface area (p < 0.001) were jointly associated with warfarin dose requirements, and together explained 33% of the variability in warfarin dose requirements among African-Americans. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 18034618-7 2007 Multiple regression analysis revealed that CYP2C9 genotype (p = 0.015), age (p < 0.001) and body surface area (p < 0.001) were jointly associated with warfarin dose requirements, and together explained 33% of the variability in warfarin dose requirements among African-Americans. Warfarin 234-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 18034618-8 2007 DISCUSSION: Our data suggest that CYP2C9 genotype, age and body size are important determinants of warfarin dose requirements in African-Americans. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 18034619-0 2007 Allelic variants in the CYP2C9 and VKORC1 loci and interindividual variability in the anticoagulant dose effect of warfarin in Italians. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 18034619-3 2007 We investigated the influence of variants of the VKORC1 and CYP2C9 loci on the mean weekly warfarin dose (MWWD) required to reach stabilized therapeutic international normalized ratio, in order to confirm and to estimate the contribution of common genetic variability of these two genes in an Italian population and to search for novel rare VKORC1 alleles. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 18040121-0 2007 Severe phenytoin toxicity in a CYP2C9*3*3 homozygous mutant from India. Phenytoin 7-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 17517049-7 2007 Recombinant CYP2C9, CYP2C18 and CYP2C19 catalysed all hydroxylation pathways, whereas CYP2C8 formed only 6beta-OHGz and 7beta-OHGz. 6beta-ohgz 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17517049-7 2007 Recombinant CYP2C9, CYP2C18 and CYP2C19 catalysed all hydroxylation pathways, whereas CYP2C8 formed only 6beta-OHGz and 7beta-OHGz. 7beta-ohgz 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17517049-8 2007 CONCLUSION: Taken together, the results indicate that CYP2C9 is the major contributor to Gz metabolic clearance, although CYP2C19 may also be involved in MeOH-Gz formation (the major metabolic pathway). gz 89-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17646413-9 2007 The decrease in voriconazole exposure was probably due to the induction of CYP2C19 and CYP2C9 by ritonavir. Voriconazole 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 17646413-9 2007 The decrease in voriconazole exposure was probably due to the induction of CYP2C19 and CYP2C9 by ritonavir. Ritonavir 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 17925902-6 2007 Similarly, genetic testing for variants in cytochrome CYP2C9 and vitamin K epoxide reductase could inform decisions about prescribing warfarin. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 18958736-3 2007 Both human CYP 2C9 and rat CYP 2C11 metabolize TA to a reactive metabolite that was reported to bind exclusively to these enzymes. Ticrynafen 47-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-18 17979511-3 2007 In humans, functionally relevant polymorphisms, which may significantly modulate epoxyeicosatrienoic acid levels in vivo, have been identified in the genes encoding CYP2J2, CYP2C8, CYP2C9 and soluble epoxide hydrolase. epoxyeicosatrienoic acid 81-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 17898651-1 2007 Cytochrome P450 2C9 (CYP4502C9) genotyping is useful in dosage adjustments for several critical drug therapies, including warfarin. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 17898651-1 2007 Cytochrome P450 2C9 (CYP4502C9) genotyping is useful in dosage adjustments for several critical drug therapies, including warfarin. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-30 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Vitamin K 239-248 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-83 17955831-4 2007 CYP2C9 and VKORC1 account for up to half the variability in vitamin K antagonists requirements and incorporating genotying data for these two genes into dosing algorithms could lead to a safer anticoagulation therapy. Vitamin K 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Vitamin K 239-248 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Warfarin 262-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-83 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Warfarin 262-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Acenocoumarol 272-285 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-83 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Acenocoumarol 272-285 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Phenprocoumon 287-300 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-83 17955831-1 2007 Single nucleotide polymorphisms (SNPs) in genes encoding cytochrome P450 enzyme 2C9 (CYP2C9) and Vitamin K epoxide reductase subunit I (VKORC1) make a significant contribution to the inter-individual variability in the maintenance dose of vitamin K antagonists (warfarin, acenocoumarol, phenprocoumon). Phenprocoumon 287-300 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17591676-3 2007 DHEA treatment elevated the expression and activities of CYP3A4, CYP2C9, CYP2C19, and CYP2B6 in primary cultures of human hepatocytes. Dehydroepiandrosterone 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17696334-4 2007 The model compound for the study is the nonsteroidal anti-inflammatory drug celecoxib, a COX-2 selective inhibitor and known CYP2C9 substrate. Celecoxib 76-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 17721328-7 2007 CYP2C9*2 C/T, CYP2C9*3 A/C, VKORC1 (-1639) G/A genotyping might be necessary for patients with Factor V Leiden and/or prothrombin G2021A mutation before warfarin anticoagulant therapy. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17960328-0 2007 Effective virtual screening protocol for CYP2C9 ligands using a screening site constructed from flurbiprofen and S-warfarin pockets. Flurbiprofen 96-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 17576809-5 2007 Human CYP2C9 metabolized esfenvalerate but not deltamethrin. fenvalerate 25-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 17960328-0 2007 Effective virtual screening protocol for CYP2C9 ligands using a screening site constructed from flurbiprofen and S-warfarin pockets. Sulfur 113-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 17960328-0 2007 Effective virtual screening protocol for CYP2C9 ligands using a screening site constructed from flurbiprofen and S-warfarin pockets. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 17960328-1 2007 An effective virtual screening protocol was developed against an extended active site of CYP2C9, which was derived from X-ray structures complexed with flubiprofen and S-warfarin. Flurbiprofen 152-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 17960328-1 2007 An effective virtual screening protocol was developed against an extended active site of CYP2C9, which was derived from X-ray structures complexed with flubiprofen and S-warfarin. Warfarin 168-178 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 17542019-2 2007 Probe substrates were phenacetin, diclofenac, S-mephenytoin, and dextromethorphan for CYP1A2, CYP2C9, CYP2C19, and CYP2D6, respectively. Dextromethorphan 65-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 17542019-2 2007 Probe substrates were phenacetin, diclofenac, S-mephenytoin, and dextromethorphan for CYP1A2, CYP2C9, CYP2C19, and CYP2D6, respectively. Phenacetin 22-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 17496208-1 2007 The elimination process of the endothelin receptor antagonist bosentan (Tracleer) in humans is entirely dependent on metabolism mediated by two cytochrome P450 (P450) enzymes, i.e., CYP3A4 and CYP2C9. Bosentan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 19356039-1 2007 Three neurotransmitters, namely adrenaline, serotonin and tryptamine inhibit the in vitro activity of several cytochrome P450 (CYP) isozymes (CYP1A2, CYP2C9, CYP2D6 and CYP3A). Epinephrine 32-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 19356039-1 2007 Three neurotransmitters, namely adrenaline, serotonin and tryptamine inhibit the in vitro activity of several cytochrome P450 (CYP) isozymes (CYP1A2, CYP2C9, CYP2D6 and CYP3A). Serotonin 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 19356039-1 2007 Three neurotransmitters, namely adrenaline, serotonin and tryptamine inhibit the in vitro activity of several cytochrome P450 (CYP) isozymes (CYP1A2, CYP2C9, CYP2D6 and CYP3A). tryptamine 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 19356039-4 2007 However, the effect became negligible when adrenaline concentration was decreased to 100 microM: whereas a high inhibitory effect was observed in CYP2D6, CYP2C9 and CYP3A4 enzyme activities, the NADPH reductase activity remains unchanged. Epinephrine 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 17827781-5 2007 A three-dimensional model for docking between EXP-3174 and CYP2C9 indicated that the chloro group of EXP-3174 is oriented to a hydrophobic pocket in the CYP2C9 active site, indicating that the lipophilicity of the group present in ARBs at the position corresponding to that of the hydroxyisopropyl group in olmesartan is important in inhibiting CYP2C9 activity. olmesartan 307-317 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 17572144-5 2007 METHODS AND RESULTS: FMD of the radial artery was determined in 12 healthy volunteers by high-resolution ultrasound and analyzed before and after intra-arterial infusion of sulfaphenazole, a specific CYP 2C9 inhibitor, L-NMMA (NO synthase inhibitor) and co-infusion of both. Sulfaphenazole 173-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 200-207 17580253-2 2007 Fluconazole inhibits cytochrome P450 2C9, which is involved with the activation of CY, and so might provide protection from CY-related toxicities. Cyclophosphamide 83-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 17580253-2 2007 Fluconazole inhibits cytochrome P450 2C9, which is involved with the activation of CY, and so might provide protection from CY-related toxicities. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 17496169-5 2007 A strong association was found between genetic polymorphisms in six genes, including VKORC1, CYP2C9, PROC, EPHX1, GGCX, and ORM1, and interindividual variability in the anticoagulant effect of warfarin; the strongest predictors were VKORC1 and CYP2C9. Warfarin 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 17496169-5 2007 A strong association was found between genetic polymorphisms in six genes, including VKORC1, CYP2C9, PROC, EPHX1, GGCX, and ORM1, and interindividual variability in the anticoagulant effect of warfarin; the strongest predictors were VKORC1 and CYP2C9. Warfarin 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 244-250 17580253-2 2007 Fluconazole inhibits cytochrome P450 2C9, which is involved with the activation of CY, and so might provide protection from CY-related toxicities. Cyclophosphamide 124-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 17580253-8 2007 These data support the hypothesis that fluconazole, when coadministered with CY, decreases CY-related toxicities by inhibiting cytochrome P450 2C9 metabolism. Fluconazole 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-146 17580253-8 2007 These data support the hypothesis that fluconazole, when coadministered with CY, decreases CY-related toxicities by inhibiting cytochrome P450 2C9 metabolism. Cyclophosphamide 77-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-146 17332144-1 2007 CYP2C9 is one of the major drug-metabolizing enzymes, and it is involved in the oxidative metabolism of approximately 10% of clinically important drugs, among which some, such as the anticoagulant warfarin, have a narrow therapeutic index. Warfarin 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17298483-0 2007 Influence of CYP2C9 and CYP2C19 genetic polymorphisms on pharmacokinetics of gliclazide MR in Chinese subjects. Gliclazide 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 17298483-1 2007 AIMS: To investigate the influence of CYP2C9 and CYP2C19 genetic polymorphisms on the pharmacokinetics of gliclazide modified release (MR) in healthy Chinese subjects. Gliclazide 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 17298483-11 2007 CONCLUSIONS: The pharmacokinetics of gliclazide MR are affected mainly by CYP2C19 genetic polymorphism instead of CYP2C9 genetic polymorphism. Gliclazide 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 17446262-4 2007 In the present study with CYP2C9, cyt b5 exerted complex actions upon P450 oxidative reactions by affecting the rate of metabolite formation, the consumption of NADPH by cytochrome P450 reductase, and uncoupling of the reaction cycle to hydrogen peroxide and water. NADP 161-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 17446262-4 2007 In the present study with CYP2C9, cyt b5 exerted complex actions upon P450 oxidative reactions by affecting the rate of metabolite formation, the consumption of NADPH by cytochrome P450 reductase, and uncoupling of the reaction cycle to hydrogen peroxide and water. Hydrogen Peroxide 237-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 17446262-4 2007 In the present study with CYP2C9, cyt b5 exerted complex actions upon P450 oxidative reactions by affecting the rate of metabolite formation, the consumption of NADPH by cytochrome P450 reductase, and uncoupling of the reaction cycle to hydrogen peroxide and water. Water 259-264 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 17446262-7 2007 The major effects of cyt b5 are hypothesized to result from a cyt b5-induced conformational change in CYP2C9 that results in an increased collision frequency between the iron-oxygen species (Cpd I) and the substrate, and a decrease in the oxidase activity. Iron 170-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 17446262-7 2007 The major effects of cyt b5 are hypothesized to result from a cyt b5-induced conformational change in CYP2C9 that results in an increased collision frequency between the iron-oxygen species (Cpd I) and the substrate, and a decrease in the oxidase activity. Oxygen 175-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 23055851-6 2007 Ibuprofen is primarily metabolized by cytochrome P450 (CYP) 2C9. Ibuprofen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-63 23055851-7 2007 The immaturity of neonatal biotransformation pathways has a pronounced effect on the pharmacokinetic parameters of ibuprofen, particularly because CYP2C9 enzyme activity is known to be very low at birth and to increase rapidly over the first several days of life. Ibuprofen 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 23055851-10 2007 Factors such as gestational age and CYP2C9 polymorphism may affect ibuprofen metabolism and therefore optimal dosing, but further clinical investigation is needed in these areas. Ibuprofen 67-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 17510308-0 2007 Estimation of warfarin maintenance dose based on VKORC1 (-1639 G>A) and CYP2C9 genotypes. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 17510308-1 2007 BACKGROUND: CYP2C9 polymorphisms are associated with decreased S-warfarin clearance and lower maintenance dosage. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17510308-6 2007 RESULTS: Eighty percent of CYP2C9*1/*1 patients stabilized on <4.0 mg/day warfarin had at least 1 VKORC1 -1639A allele. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 17510308-9 2007 A model including VKORC1 and CYP2C9 genotypes, age, sex, and body weight accounted for 61% of the variance in warfarin daily maintenance dose. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 17510308-12 2007 VKORC1 and CYP2C9 genotypes, age, sex, and body weight account for the majority of variance in warfarin dose among our study population. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). coumarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-37 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). coumarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). Acenocoumarol 120-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-37 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). Acenocoumarol 120-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-37 17611042-4 2007 Secondly, SNPs of cytochrome P450 2C9 (CYP2C9) gene have been shown to decrease the catabolism of coumarin derivatives (acenocoumarol, warfarin). Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 17611042-7 2007 Finally, the combined analysis of VKORC1 and CYP2C9 SNPs with age may account for more than 50% of the individual variability of the warfarin maintenance dosage. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 18240910-4 2007 Recently, polymorphisms in two genes, cytochrome P450 2C9 and vitamin K epoxide reductase complex 1, have been shown to affect warfarin"s pharmacogenomics and pharmacodynamics, respectively. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-57 17433521-4 2007 Curcumin inhibited CYP1A2 (IC(50), 40.0 microM), CYP3A4 (IC(50), 16.3 microM), CYP2D6 (IC(50), 50.3 microM), CYP2C9 (IC(50), 4.3 microM) and CYP2B6 (IC(50), 24.5 microM). Curcumin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 17516991-0 2007 Inhibitory effect of valproic acid on cytochrome P450 2C9 activity in epilepsy patients. Valproic Acid 21-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-57 17516991-4 2007 The aim of this study was to evaluate the inhibitory effect of valproic acid on cytochrome P450 2C9 (CYP2C9) activity by using losartan oxidation as a probe in epilepsy patients. Valproic Acid 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-99 17516991-4 2007 The aim of this study was to evaluate the inhibitory effect of valproic acid on cytochrome P450 2C9 (CYP2C9) activity by using losartan oxidation as a probe in epilepsy patients. Valproic Acid 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 17516991-4 2007 The aim of this study was to evaluate the inhibitory effect of valproic acid on cytochrome P450 2C9 (CYP2C9) activity by using losartan oxidation as a probe in epilepsy patients. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-99 17516991-4 2007 The aim of this study was to evaluate the inhibitory effect of valproic acid on cytochrome P450 2C9 (CYP2C9) activity by using losartan oxidation as a probe in epilepsy patients. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 17516991-7 2007 Losartan and E3174, the CYP2C9-derived carboxylic acid metabolite of losartan in 8 hr urine were assayed by using high pressure liquid chromatography. Carboxylic Acids 39-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 17516991-7 2007 Losartan and E3174, the CYP2C9-derived carboxylic acid metabolite of losartan in 8 hr urine were assayed by using high pressure liquid chromatography. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 17516991-10 2007 The results suggest an inhibitory effect of valproic acid on CYP2C9 enzyme activity in epilepsy patients at steady state. Valproic Acid 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 17332144-3 2007 We found a new sequence variation in exon 7 of the CYP2C9 gene (1060G>A) resulting in a substitution of acidic amino acid glutamate to basic lysine (E354K) when translated. amino acid glutamate 114-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 17332144-3 2007 We found a new sequence variation in exon 7 of the CYP2C9 gene (1060G>A) resulting in a substitution of acidic amino acid glutamate to basic lysine (E354K) when translated. Lysine 144-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 17332144-4 2007 The allele, designated CYP2C9(*)24, was present in heterozygous state in one warfarin-treated patient. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 17332144-9 2007 These results suggest that CYP2C9.24 may be improperly folded, both in yeast and mammalian cells, resulting in improper heme incorporation and rapid intracellular degradation. Heme 120-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 17468864-10 2007 Furthermore, BTC was found to be a potent inhibitor of CYP3A4 and CYP2C9 activity in vitro. Benzethonium 13-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 17612953-11 2007 Tienilic acid is activated by and covalently bound to CYP2C9, and the neoantigens thus formed activate immune systems, resulting in the formation of an autoantibodydirected against CYP2C9, named anti-liver/kidney microsomal autoantibody type 2, whereas the pathological role of the autoantibodies in drug-induced hepatitis remains largely unknown. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17612953-11 2007 Tienilic acid is activated by and covalently bound to CYP2C9, and the neoantigens thus formed activate immune systems, resulting in the formation of an autoantibodydirected against CYP2C9, named anti-liver/kidney microsomal autoantibody type 2, whereas the pathological role of the autoantibodies in drug-induced hepatitis remains largely unknown. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 17596671-0 2007 A case report of a patient carrying CYP2C9*3/4 genotype with extremely low warfarin dose requirement. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 17596671-1 2007 We report a case of intolerance to warfarin dosing due to impaired drug metabolism in a patient with CYP2C9*3/*4. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 17562299-0 2007 Contributions of CYP2C9/CYP2C19 genotypes and drug interaction to the phenytoin treatment in the Korean epileptic patients in the clinical setting. Phenytoin 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 16969365-0 2007 Genetic variation at the CYP2C locus and its association with torsemide biotransformation. Torsemide 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-30 16969365-2 2007 Torsemide total oral clearance was linearly associated with the number of CYP2C9(*)3 alleles (geometric mean: 59, 40 and 20 ml/min in carriers of no, one and two alleles) and so were the methyl- and ring-hydroxylation but not the carboxylation clearance. Torsemide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 16969365-5 2007 Torsemide total oral clearance was predictable with r(2)=82.1% using plasma concentrations at 0.5, 1, 2 and 24 h. In conclusion, torsemide"s biotransformation strongly depended on the CYP2C9(*)3 variant but no other. Torsemide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 16969365-5 2007 Torsemide total oral clearance was predictable with r(2)=82.1% using plasma concentrations at 0.5, 1, 2 and 24 h. In conclusion, torsemide"s biotransformation strongly depended on the CYP2C9(*)3 variant but no other. Torsemide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 17562299-2 2007 The CYP2C9 polymorphism is a wellknown major genetic factor responsible for phenytoin metabolism. Phenytoin 76-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17562299-4 2007 Using a multiple regression model for evaluation of the CYP2C9 and CYP2C19 genotypes, together with other non-genetic variables, we explained 39.6% of the variance in serum phenytoin levels. Phenytoin 173-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 17562299-5 2007 Incorporation of genotyping for CYP2C9 and CYP2C19 into a clinical practice may be of some help in the determination of phenytoin dosage. Phenytoin 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 17442686-1 2007 This study assessed the effects of clopidogrel, a CYP 2C9 inhibitor, on fluvastatin pharmacokinetics in healthy volunteers. Clopidogrel 35-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-57 17504998-4 2007 The purposes of this study were to evaluate the effect of genetic variants of CYP2C9 and CYP2C19 on the pharmacokinetics of indisulam and on clinical outcome and to assess the need for pharmacogenetically guided dose adaptation. N-(3-chloro-7-indolyl)-1,4-benzenedisulphonamide 124-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 17361128-3 2007 Clopidogrel"s activation involves two sequential steps by CYP3A, CYP1A2, CYP2C9, CYP2C19, and/or CYP2B6. Clopidogrel 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 17513950-9 2007 Treatment of endothelial cells with lovastatin increased CYP2C9 expression. Lovastatin 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 17513950-10 2007 After 96 hours of treatment, fluvastatin and lovastatin clearly increased CYP2C9 protein level. Fluvastatin 29-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 17513950-10 2007 After 96 hours of treatment, fluvastatin and lovastatin clearly increased CYP2C9 protein level. Lovastatin 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 17442935-7 2007 The enzymes CYP2C9, CYP2C8, and myeloperoxidase catalyzed the conversion of sulfamethoxazole to sulfamethoxazole hydroxylamine. Sulfamethoxazole 76-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 17442935-7 2007 The enzymes CYP2C9, CYP2C8, and myeloperoxidase catalyzed the conversion of sulfamethoxazole to sulfamethoxazole hydroxylamine. sulfamethoxazole hydroxylamine 96-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 19802360-0 2007 Influence of CYP2C9 Genotype on warfarin dose among African American and European Americans. Warfarin 32-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 19802360-4 2007 Herein we report the influence of CYP2C9 genotype on warfarin dose among European American and African American patients. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 19802360-16 2007 Warfarin dose was significantly related to CYP2C9 genotype (p<0.0001) both in univariate and multivariate analyses. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 19802360-20 2007 CYP2C9 genotype predicts warfarin dose in European Americans, but not in African Americans. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17479200-0 2007 Any effect of CYP2C9 variants on warfarin clearance in Chinese patients? Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. chlorethylclonidine 240-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. Fluvoxamine 245-256 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. Fluvastatin 258-264 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 17307149-2 2007 In order to gain insights for developing personalized drugs, the 3D (dimensional) structure of CYP2C19 has been developed based on the crystal structure of CYP2C9 (PDB code 1R90), and its structure-activity relationship with the ligands of CEC, Fluvoxamine, Lescol, and Ticlopidine investigated through the structure-activity relationship approach. Ticlopidine 270-281 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 17054666-0 2007 Evaluation of flurbiprofen urinary ratios as in vivo indices for CYP2C9 activity. Flurbiprofen 14-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17301738-5 2007 CYP2C9 genotype and age were identified as predictors for S-warfarin clearance, and VKORC1 genotype as a predictor for warfarin sensitivity. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17220242-1 2007 The cytochrome P450 (P450) enzymes CYP2C8, CYP2C9, and CYP2J2 metabolize arachidonic acid to epoxyeicosatrienoic acids, which are known to be vital in regulation of vascular tone and cardiovascular homeostasis. Arachidonic Acid 73-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 17220242-1 2007 The cytochrome P450 (P450) enzymes CYP2C8, CYP2C9, and CYP2J2 metabolize arachidonic acid to epoxyeicosatrienoic acids, which are known to be vital in regulation of vascular tone and cardiovascular homeostasis. epoxyeicosatrienoic acids 93-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 17251307-5 2007 Therefore, the three nitrogenous ligands aniline, imidazole, and triazole were used as binding spectra standards with purified human CYP3A4 and CYP2C9, because their small size should not present any steric limitations in their accessing the heme prosthetic group. aniline 41-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 17251307-5 2007 Therefore, the three nitrogenous ligands aniline, imidazole, and triazole were used as binding spectra standards with purified human CYP3A4 and CYP2C9, because their small size should not present any steric limitations in their accessing the heme prosthetic group. imidazole 50-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 17251307-5 2007 Therefore, the three nitrogenous ligands aniline, imidazole, and triazole were used as binding spectra standards with purified human CYP3A4 and CYP2C9, because their small size should not present any steric limitations in their accessing the heme prosthetic group. Triazoles 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 17377945-0 2007 Study of recombinant cytochrome P450 2C9 activity with diclofenac by MEKC. Diclofenac 55-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 17377945-2 2007 The aim of this paper is to demonstrate the applicability of CE for the determination of the enzymatic activity of CYP2C9 with diclofenac as a probe substrate. Diclofenac 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 17603219-4 2007 The CYP2C9SV protein showed a typical reduced CO-difference spectrum, indicating that the translated protein binds a heme moiety. Heme 117-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17391071-6 2007 There were statistically significant differences in warfarin dose between the CYP2C9*1/*1 (4.3 +/- 1.6 mg/day; p < 0.001) and those with the other two genotypes including CYP2C9*1/*3 and CYP2C9*3/*4 (2.7 +/- 0.9 mg/day). Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 16983400-4 2007 The most important genes affecting the pharmacokinetic and pharmacodynamic parameters of warfarin are CYP2C9 (cytochrome P(450) 2C9) and VKORC1 (vitamin K epoxide reductase complex subunit 1). Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 16983400-4 2007 The most important genes affecting the pharmacokinetic and pharmacodynamic parameters of warfarin are CYP2C9 (cytochrome P(450) 2C9) and VKORC1 (vitamin K epoxide reductase complex subunit 1). Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-131 17391071-6 2007 There were statistically significant differences in warfarin dose between the CYP2C9*1/*1 (4.3 +/- 1.6 mg/day; p < 0.001) and those with the other two genotypes including CYP2C9*1/*3 and CYP2C9*3/*4 (2.7 +/- 0.9 mg/day). Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 17391071-6 2007 There were statistically significant differences in warfarin dose between the CYP2C9*1/*1 (4.3 +/- 1.6 mg/day; p < 0.001) and those with the other two genotypes including CYP2C9*1/*3 and CYP2C9*3/*4 (2.7 +/- 0.9 mg/day). Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 17303175-4 2007 Human CYP2C9-Arg expressed in the microsomes of human B lymphoblastoid cells efficiently catalyzed the 11-hydroxylation of Delta(8)-THC (7.60 nmol/min/nmol CYP), Delta(9)-THC (19.2 nmol/min/nmol CYP) and CBN (6.62 nmol/min/nmol CYP). Arginine 13-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 17311370-3 2007 We have used analogues of the prototypical CYP2C9 heteroactivator dapsone to validate a simple docking method that can be used to predict heteroactivators based on ligand binding location in a P450 crystal structure. Dapsone 66-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 17311370-5 2007 One of the top-scoring compounds identified was verified to be a CYP2C9 heteroactivator in vitro, and it possessed activity similar to dapsone. Dapsone 135-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17303175-4 2007 Human CYP2C9-Arg expressed in the microsomes of human B lymphoblastoid cells efficiently catalyzed the 11-hydroxylation of Delta(8)-THC (7.60 nmol/min/nmol CYP), Delta(9)-THC (19.2 nmol/min/nmol CYP) and CBN (6.62 nmol/min/nmol CYP). delta-8-tetrahydrocannabinol 123-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 17110455-1 2007 CYP2C9 and VKORC1 genetic variants are associated with low and intermediate warfarin dose requirements, but markers of high doses are less well characterized. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17303175-4 2007 Human CYP2C9-Arg expressed in the microsomes of human B lymphoblastoid cells efficiently catalyzed the 11-hydroxylation of Delta(8)-THC (7.60 nmol/min/nmol CYP), Delta(9)-THC (19.2 nmol/min/nmol CYP) and CBN (6.62 nmol/min/nmol CYP). Dronabinol 162-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 17322161-5 2007 Specifically, cranberry juice may inhibit the activity of CYP2C9, the primary isoenzyme involved in the metabolism of S-warfarin. Warfarin 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 17456829-6 2007 RESULTS: Of genetic factors evaluated in the model, CYP2C9 and VKORC1 each contributed substantially to dose variability, and together with clinical factors explained 56% of the individual variability in stable warfarin dose. Warfarin 211-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 17456829-8 2007 CONCLUSION: The importance of CYP2C9 and VKORC1 to patient-specific dose of warfarin has been confirmed, while polymorphisms of gamma carboxylase, factor VII and Apo E genes did not substantially contribute to predictive models for stable warfarin dose. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 17324110-0 2007 Warfarin and cytochrome P450 2C9 genotype: possible ethnic variation in warfarin sensitivity. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-32 17305793-0 2007 CYP2C9 variant modifies blood pressure-lowering response to losartan in Type 1 diabetic patients with nephropathy. Losartan 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17577464-0 2007 Warfarin metabolism and anticoagulant effect: a prospective, observational study of the impact of CYP2C9 genetic polymorphism in the presence of drug-disease and drug-drug interactions. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 17577464-1 2007 BACKGROUND: Cytocbrome P450 (CYP) 2C9 polymorphism affects the warfarin dosage requirement in stable outpatients. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 17577464-3 2007 OBJECTIVE: The aim of this study was to examine the effects of CYP2C9 genetic polymorphism on warfarin dosage requirements in patients with severe comorbid conditions and those treated with medications that potentially interact with warfarin. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 17577464-3 2007 OBJECTIVE: The aim of this study was to examine the effects of CYP2C9 genetic polymorphism on warfarin dosage requirements in patients with severe comorbid conditions and those treated with medications that potentially interact with warfarin. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 17577464-14 2007 CONCLUSIONS: In this study population of patients with severe comorbid conditions and those treated with medications that potentially interact with warfarin, CYP2C9 *1/*3 genotype, older age, CHF, and the use of antibiotics were associated with lower warfarin dosage requirements. Warfarin 251-259 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 17577464-15 2007 The CYP2C9*1/*3 genotype, compared with CYP2C9 *1/*1, was associated with 33% lower mean warfarin dosage requirements and higher INR values, which were higher than the upper therapeutic range of INR (ie, 3). Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17577464-15 2007 The CYP2C9*1/*3 genotype, compared with CYP2C9 *1/*1, was associated with 33% lower mean warfarin dosage requirements and higher INR values, which were higher than the upper therapeutic range of INR (ie, 3). Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 17577464-16 2007 Genetic CYP2C9 polymorphism contributed to the variability in warfarin dosage requirements in the presence of drug-disease and drug-drug interactions. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 17048007-7 2007 A multiple regression model with VKORC1, CYP2C9, PROC and the non-genetic predictors age, bodyweight, drug interactions and indication for treatment jointly accounted for 62% of variance in warfarin dose. Warfarin 190-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 17324110-3 2007 The aim was to evaluate the influence of two CYP2C9 functional polymorphisms (*2 and *3) on warfarin dose in African-Americans, an unstudied population and Caucasians, and also to assess the effect of these polymorphisms on anticoagulation response after accounting for nongenetic factors and genetic factors that might also impact the dose-response relationship of warfarin. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 29793299-2 2007 : A warfarin-dosing model in Asians that uses single nucleotide polymorphisms in vitamin K epoxide reductase complex and cytochrome P450 2C9. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-140 17284505-3 2007 CYP2C9 is responsible for the irreversible metabolism of ibuprofen. Ibuprofen 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17132763-5 2007 In addition, we investigated the effect of pomegranate juice on the pharmacokinetics of tolbutamide (substrate for CYP2C9) in rats. Tolbutamide 88-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 29793299-8 2007 Tham and colleagues used specific CYP2C9 and VKORC1 polymorphisms, along with clinical factors, to build an algorithm to accurately estimate warfarin doses in a multi-ethnic Asian population. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 29793302-4 2007 In particular, the variability in warfarin dose requirement is now recognized to be due, in large part, to polymorphisms in two genes: cytochrome P450 2C9 and the vitamin K epoxide reductase complex subunit 1. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-154 17304159-0 2007 Population estimation of the effects of cytochrome P450 2C9 and 2C19 polymorphisms on phenobarbital clearance in Japanese. Phenobarbital 86-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-59 17304159-1 2007 A nonlinear mixed-effect modeling (NONMEM) program was used to evaluate the effects of cytochrome P450 (CYP) 2C9 and CYP2C19 polymorphisms on the phenobarbital (PB) population clearance for Japanese epileptics. Phenobarbital 146-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-112 17304159-5 2007 The total clearance of PB decreased by 48% in patients with CYP2C9*1/*3 genotype in comparison with those with CYP2C9*1/*1 genotype (P < 0.001). Phenobarbital 23-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 17304159-5 2007 The total clearance of PB decreased by 48% in patients with CYP2C9*1/*3 genotype in comparison with those with CYP2C9*1/*1 genotype (P < 0.001). Phenobarbital 23-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 17304159-7 2007 To our knowledge, this is the first report to demonstrate that the CYP2C9 genotype affects the PB metabolism in routine care, but the results should be further verified in other ethnic populations. Phenobarbital 95-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 17375979-8 2007 CYP2C19 contributes to the metabolism of diazepam and phenytoin, the latter drug also representing a substrate of CYP2C9, with its predominant variants being defined as *2 and *3. Diazepam 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 16822276-9 2007 CONCLUSIONS: Coadministration of potent inhibitors of CYP3A4/5 and CYP2D6, but not CYP2C9, resulted in a moderate increase in lasofoxifene exposure. Lasofoxifene 126-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 17201456-6 2007 CYP2C8 and CYP3A4 are the main isoenzymes catalysing biotransformation of pioglitazone (as with troglitazone), whereas rosiglitazone is metabolised by CYP2C9 and CYP2C8. Rosiglitazone 119-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 17375979-8 2007 CYP2C19 contributes to the metabolism of diazepam and phenytoin, the latter drug also representing a substrate of CYP2C9, with its predominant variants being defined as *2 and *3. Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 17375979-9 2007 For both AEDs, there is maximally a 2-fold difference in the hepatic elimination rate (e.g. clearance) or the AUC between the extremes of EMs and PMs which, in the case of phenytoin (an AED with a narrow "therapeutic window"), would suggest a dosage reduction only for patients who are carriers of mutated alleles of both CYP2C19 and CYP2C9, a subgroup that is very rare among Caucasians (about 1% of the population) but more frequent in Asians (about 10%). Phenytoin 172-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 334-340 17167248-0 2007 Frequency of CYP2C9 genotypes among Omani patients receiving warfarin and its correlation with warfarin dose. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 17167248-0 2007 Frequency of CYP2C9 genotypes among Omani patients receiving warfarin and its correlation with warfarin dose. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 17167248-1 2007 OBJECTIVES: This study was conducted to determine the frequency of CYP2C9 alleles in Omani patients receiving warfarin and to correlate genotyping data with warfarin dosage. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 17167248-1 2007 OBJECTIVES: This study was conducted to determine the frequency of CYP2C9 alleles in Omani patients receiving warfarin and to correlate genotyping data with warfarin dosage. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 17284096-3 2007 In particular, two polymorphisms of cytochrome P450 2C9 are known to slow down the metabolism of phenytoin to a degree that increases the risk of the neurotoxic adverse effects of this drug among carriers of these polymorphisms. Phenytoin 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-55 18690851-2 2007 Consistent with in vitro data, clinical studies have demonstrated that CYP2C9 polymorphisms significantly influence warfarin pharmacokinetics by reducing (S)-warfarin metabolic clearance, consequently lowering maintenance dose requirements and increasing the risk over-anticoagulation during the initiation phase of therapy. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 18690851-2 2007 Consistent with in vitro data, clinical studies have demonstrated that CYP2C9 polymorphisms significantly influence warfarin pharmacokinetics by reducing (S)-warfarin metabolic clearance, consequently lowering maintenance dose requirements and increasing the risk over-anticoagulation during the initiation phase of therapy. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Warfarin 156-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 17708066-11 2007 Diosmin might have inhibited the microsomal CYP2C9 mediated oxidation of diclofenac sodium. Diosmin 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 17708066-11 2007 Diosmin might have inhibited the microsomal CYP2C9 mediated oxidation of diclofenac sodium. Diclofenac 73-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 17253883-8 2007 In addition to CYP3A4, repaglinide is metabolised via CYP2C8, while nateglinide metabolism also involves CYP2C9. Nateglinide 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 17020955-7 2007 In human liver microsomes, involvement of CYP2D6, CYP1A2, CYP2C9, and CYP2C19 in diphenhydramine N-demethylation was confirmed by using P450 isozyme-specific inhibitors. Diphenhydramine 81-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 17020955-7 2007 In human liver microsomes, involvement of CYP2D6, CYP1A2, CYP2C9, and CYP2C19 in diphenhydramine N-demethylation was confirmed by using P450 isozyme-specific inhibitors. Nitrogen 97-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 17020955-11 2007 In addition, large differences in the metabolic activities of CYP2D6 and those of CYP1A2, CYP2C9, and CYP2C19 could cause the individual differences in anti-allergic efficacy and the sedative effect of diphenhydramine. Diphenhydramine 202-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 17419358-5 2007 Basing on the results of the study, the authors propose an algorithm of choosing the initial warfarin dose depending on CYP2C9 genotype. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 17192504-7 2007 These data demonstrate a CYP2C9-mediated pharmacokinetic interaction with pharmacodynamic, clinically relevant consequences, which might require warfarin dose adjustment. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 17344941-4 2007 Losartan metabolism has been suggested as a marker for determination of CYP2C9 activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 17419358-0 2007 [The influence of CYP2C9 genetic polymorphism on the pharmacokinetics and pharmacodynamics of warfarin in patients with constant atrial fibrillation]. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 17419358-1 2007 CYP2C9 is the main enzyme participating in warfarin metabolism, of which genetic polymorphism is typical. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17419358-2 2007 The aim of the study was to investigate the influence of having allelic variants CYP2C9*2 and CYP2C9*3 on the pharmacokinetics, dosage regimen, and the rate of hemorrhage in patients with constant atrial fibrillation taking warfarin. Warfarin 224-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 17419358-4 2007 It was shown that in patients with CYP2C9*2 and CYP2C9*3 the clearance of warfarin and its dose were lower, while the episodes of excessive hypocoagulation and hemorrhage associated with warfarin were more frequent than in patients without these allelic variants. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 17419358-4 2007 It was shown that in patients with CYP2C9*2 and CYP2C9*3 the clearance of warfarin and its dose were lower, while the episodes of excessive hypocoagulation and hemorrhage associated with warfarin were more frequent than in patients without these allelic variants. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 17419358-4 2007 It was shown that in patients with CYP2C9*2 and CYP2C9*3 the clearance of warfarin and its dose were lower, while the episodes of excessive hypocoagulation and hemorrhage associated with warfarin were more frequent than in patients without these allelic variants. Warfarin 187-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 17419358-4 2007 It was shown that in patients with CYP2C9*2 and CYP2C9*3 the clearance of warfarin and its dose were lower, while the episodes of excessive hypocoagulation and hemorrhage associated with warfarin were more frequent than in patients without these allelic variants. Warfarin 187-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 17049586-0 2007 Genotypes of vitamin K epoxide reductase, gamma-glutamyl carboxylase, and cytochrome P450 2C9 as determinants of daily warfarin dose in Japanese patients. Warfarin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-93 17397249-0 2007 The influence of NSAIDs on coumarin sensitivity in patients with CYP2C9 polymorphism after total hip replacement surgery. coumarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 17397249-1 2007 OBJECTIVE: To determine the influence of NSAIDs on the international normalized ratio (INR) in patients with cytochrome P450 (CYP)-2C9 enzyme variants starting acenocoumarol (an oral coumarin) therapy during the first 7 days after total hip replacement surgery. Acenocoumarol 160-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-134 17672075-17 2007 Episodes of MHO rise > 5.0 in warfarin therapy were observed in 50% carriers of CYP2C9*3 and in none homozygous carriers of CYP2C9*1 (p = 0.024). Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 17672075-17 2007 Episodes of MHO rise > 5.0 in warfarin therapy were observed in 50% carriers of CYP2C9*3 and in none homozygous carriers of CYP2C9*1 (p = 0.024). Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 17672075-18 2007 CYP2C9*3 patients needed lower maintenance doses of warfarin, in CYP2C9*1 and CYP2C9*2 patients anticoagulant doses were comparable. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 17672075-21 2007 Determination of CYP2C9 genotype in APS patients before treatment with oral anticoagulants may help in planning individual policy and in reducing the risk of warfarin overdosage at the start of therapy. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 17963417-6 2007 Multiple studies indicated that the CYP2C9 *3 allele (Ile359Leu polymorphism) was associated with decreased clearance of sulfonylurea drugs. Sulfonylurea Compounds 121-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 17963417-7 2007 Supporting this, one study reported an increased insulin secretion in CYP2C9*3 allele carriers when using the sulfonylurea agent glyburide. Glyburide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 17963417-8 2007 The CYP2C9*3 allele was also associated with a decreased clearance of meglitinides, whereas the CYP2C8*3 (Arg139Lys; Lys399Arg) variant increased the clearance of meglitinides. meglitinide 70-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17963417-8 2007 The CYP2C9*3 allele was also associated with a decreased clearance of meglitinides, whereas the CYP2C8*3 (Arg139Lys; Lys399Arg) variant increased the clearance of meglitinides. meglitinide 163-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17279482-6 2007 Among them, tanshinone IIA and cryptotanshinone are found to be potent inhibitors to CYP1A2, while artemisinin is a marginal inhibitor to CYP1A2 and glycyrrhetic acid is a weak inhibitor to CYP2C9. tanshinone 12-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 17279482-6 2007 Among them, tanshinone IIA and cryptotanshinone are found to be potent inhibitors to CYP1A2, while artemisinin is a marginal inhibitor to CYP1A2 and glycyrrhetic acid is a weak inhibitor to CYP2C9. cryptotanshinone 31-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 17279482-6 2007 Among them, tanshinone IIA and cryptotanshinone are found to be potent inhibitors to CYP1A2, while artemisinin is a marginal inhibitor to CYP1A2 and glycyrrhetic acid is a weak inhibitor to CYP2C9. Glycyrrhetinic Acid 149-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 17049586-4 2007 We genotyped eleven representative single nucleotide polymorphisms (SNPs) in the three genes involved in vitamin K cycle and the 42613A>C SNP in CYP2C9, known as CYP2C93, and then examined an association of these genotypes with warfarin maintenance doses (mean+/-SD=2.96+/-1.06 mg/day). Warfarin 231-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 17049586-5 2007 We found an association of effective warfarin dose with the -1639G>A (p=0.004) and 3730G>A genotypes (p=0.006) in VKORC1, the 8016G>A genotype in GGCX (p=0.022), and the 42613A>C genotype in CYP2C9 (p=0.015). Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 17049586-7 2007 The contribution to inter-individual variation in warfarin dose was 5.9% for VKORC1 -1639G>A, 5.2% for CYP2C9 42613A>C, and 4.6% for GGCX 8016G>A. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 17097122-9 2006 The CYP2C9 inhibitor sulphaphenazole (10 microM) protected from diclofenac-induced cell injury and prevented increases in [Ca(2+)](c), while it had no effect on the dissipation of the DeltaPsi(m). Sulfaphenazole 21-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 17010942-3 2006 The P450 M-2C enzyme expressed in yeast cells catalyzed p-methylhydroxylation of only tolbutamide among four substrates tested, paclitaxel as a CYP2C8 substrate, diclofenac and tolbutamide as CYP2C9 substrates and S-mephenytoin as a CYP2C19 substrate. Tolbutamide 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 17010942-5 2006 Although all of the recombinant human CYP2C8, CYP2C9 and CYP2C19 expressed in yeast cells catalyzed tolbutamide p-methylhydroxylation, the kinetic profile of CYP2C8 was most similar to that of P450 M-2C. Tolbutamide 100-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 17010942-6 2006 Tolbutamide oxidation by the marmoset liver microsomes and the recombinant P450 M-2C was inhibited most effectively by quercetin, a CYP2C8 inhibitor, followed by omeprazole, a CYP2C19 inhibitor, whereas sulfaphenazole, a CYP2C9 inhibitor, was less potent under the conditions used. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 17010942-6 2006 Tolbutamide oxidation by the marmoset liver microsomes and the recombinant P450 M-2C was inhibited most effectively by quercetin, a CYP2C8 inhibitor, followed by omeprazole, a CYP2C19 inhibitor, whereas sulfaphenazole, a CYP2C9 inhibitor, was less potent under the conditions used. Quercetin 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 17161452-3 2007 Pharmacogenetic analysis of two genes, the warfarin metabolic enzyme CYP2C9 and warfarin target enzyme, vitamin K epoxide reductase complex 1 VKORC1, confirmed their influence on warfarin maintenance dose. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 17161452-4 2007 Possession of CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, is associated with a significant decrease in the mean warfarin dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 17161452-4 2007 Possession of CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, is associated with a significant decrease in the mean warfarin dose. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 17161452-6 2007 Haplotypes based on these SNPs explain a large fraction of the interindividual variation in warfarin dose, and VKORC1 has an approximately three-fold greater effect than CYP2C9. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 17161452-7 2007 Algorithms incorporating genetic (CYP2C9 and VKORC1), demographic, and clinical factors to estimate the warfarin dosage, could potentially minimize the risk of over dose during warfarin induction. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 17161452-7 2007 Algorithms incorporating genetic (CYP2C9 and VKORC1), demographic, and clinical factors to estimate the warfarin dosage, could potentially minimize the risk of over dose during warfarin induction. Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 17097122-9 2006 The CYP2C9 inhibitor sulphaphenazole (10 microM) protected from diclofenac-induced cell injury and prevented increases in [Ca(2+)](c), while it had no effect on the dissipation of the DeltaPsi(m). Diclofenac 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 16978758-7 2006 Since the CYP2C18 content of hepatic microsomes in humans is relatively low, CYP2C9, 2C19, and 3A4 might be the main isoforms of CYP that are responsible for tributyltin and triphenyltin metabolism in the human liver. tributyltin 158-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 16978758-7 2006 Since the CYP2C18 content of hepatic microsomes in humans is relatively low, CYP2C9, 2C19, and 3A4 might be the main isoforms of CYP that are responsible for tributyltin and triphenyltin metabolism in the human liver. triphenyltin 174-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 17178259-6 2006 Cerivastatin is metabolized by CYP2C8 and CYP3A4, and fluvastatin is metabolized by CYP2C9. Fluvastatin 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 17178259-7 2006 The exposure to fluvastatin is increased by less than 2-fold by inhibitors of CYP2C9. Fluvastatin 16-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 16963489-3 2006 The most abundant isoform of this subfamily is CYP2C9, which is the major clearance pathway for the low therapeutic index drugs warfarin and phenytoin. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 16963489-3 2006 The most abundant isoform of this subfamily is CYP2C9, which is the major clearance pathway for the low therapeutic index drugs warfarin and phenytoin. Phenytoin 141-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 17042764-7 2006 A number of factors such as patient age, body weight, co-morbidity, frailty, warfarin daily dose and CYP2C9 and VKORC1 polymorphism could affect response to vitamin K and thus the rate and extent of INR reversal. Vitamin K 157-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 17111199-0 2006 Genotypes of the cytochrome p450 isoform, CYP2C9, and the vitamin K epoxide reductase complex subunit 1 conjointly determine stable warfarin dose: a prospective study. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 17111199-1 2006 BACKGROUND: Warfarin has a narrow therapeutic range and wide inter-individual dosing requirements that may be related to functional variants of genes affecting warfarin metabolism (i.e., CYP2C9) and activity (i.e., vitamin K epoxide reductase complex subunit 1-VKORC1). Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 17111199-1 2006 BACKGROUND: Warfarin has a narrow therapeutic range and wide inter-individual dosing requirements that may be related to functional variants of genes affecting warfarin metabolism (i.e., CYP2C9) and activity (i.e., vitamin K epoxide reductase complex subunit 1-VKORC1). Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 17111199-11 2006 CONCLUSION: In this large prospective study of warfarin genetic dose-determinants, carriage of a single or double CYP2C9 variant, reduced warfarin dose 18-72%, and of a VKORC1 variant by 65%. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 17111199-11 2006 CONCLUSION: In this large prospective study of warfarin genetic dose-determinants, carriage of a single or double CYP2C9 variant, reduced warfarin dose 18-72%, and of a VKORC1 variant by 65%. Warfarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 17162464-5 2006 Adefovir and its prodrug inhibited CYP2C9 at concentrations below 100 microM; inhibition by adefovir was of the uncompetitive (at the lower inhibitor concentrations) or of the competitive nature with a Ki = 420 microM. adefovir 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 16945988-0 2006 Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Valproic Acid 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 16945988-0 2006 Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Valproic Acid 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 2-propyl-4-pentenoic acid 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 4-Hydroxyvalproic acid 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16945988-2 2006 cDNA-expressed CYP2C9*1, CYP2A6, and CYP2B6 were the most active catalysts of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA formation. 5-Hydroxyvalproic acid 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16945988-9 2006 Overall, our novel findings indicate that in human hepatic microsomes, CYP2C9*1 is the predominant catalyst in the formation of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA, whereas CYP2A6 contributes partially to 3-OH-VPA formation. 2-propyl-4-pentenoic acid 128-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16945988-9 2006 Overall, our novel findings indicate that in human hepatic microsomes, CYP2C9*1 is the predominant catalyst in the formation of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA, whereas CYP2A6 contributes partially to 3-OH-VPA formation. 4-Hydroxyvalproic acid 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16945988-9 2006 Overall, our novel findings indicate that in human hepatic microsomes, CYP2C9*1 is the predominant catalyst in the formation of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA, whereas CYP2A6 contributes partially to 3-OH-VPA formation. 5-Hydroxyvalproic acid 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16945988-9 2006 Overall, our novel findings indicate that in human hepatic microsomes, CYP2C9*1 is the predominant catalyst in the formation of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA, whereas CYP2A6 contributes partially to 3-OH-VPA formation. 3-Hydroxyvalproic acid 203-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 17213583-6 2006 The activities of CYP1A2 and CYP2C9 were significantly reduced in the present of TMX. 1,3,8-trihydroxy-5-methoxyxanthone 81-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 17213583-7 2006 CONCLUSION: TMX (10 micromol/L) has significant inhibitive effect on the activities of CYP1A2 and CYP2C9, but no significant inhibitive effect on the activities of CYP2C19, CYP2E1 and CYP3A4. 1,3,8-trihydroxy-5-methoxyxanthone 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 16978661-8 2006 Incubation of PUFAs with CYP2C9 or CYP2C19 in the presence of NADPH resulted in the decrease of PUFA concentrations in the incubation mixtures. NADP 62-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 17162464-6 2006 Tenofovir and tenofovir disoproxil influenced the activity of CYP2C9, and competitive inhibition was found with Ki = 580 and 395 microM, respectively. Tenofovir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 17162464-6 2006 Tenofovir and tenofovir disoproxil influenced the activity of CYP2C9, and competitive inhibition was found with Ki = 580 and 395 microM, respectively. Tenofovir 14-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 16869821-4 2006 Results The acenocoumarol-sensitive patient was found to possess, in addition to CYP2C9*3 allele, a CYP2C9*11 allele and the VKORC1 AA diplotype which were all traced back through the parental lines. Acenocoumarol 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 16928789-3 2006 We measured the Michaelis-Menten kinetic parameters for four CYP2C9 substrates (diclofenac, (S)-warfarin, tolbutamide, and (S)-flurbiprofen) using four recombinant CYP2C9 enzyme systems (Supersomes, Baculosomes, RECO system, and in-house purified, reconstituted enzyme) to determine whether the enzyme systems exhibited kinetic differences in metabolic product formation rates under uniform experimental conditions. Diclofenac 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 16928789-3 2006 We measured the Michaelis-Menten kinetic parameters for four CYP2C9 substrates (diclofenac, (S)-warfarin, tolbutamide, and (S)-flurbiprofen) using four recombinant CYP2C9 enzyme systems (Supersomes, Baculosomes, RECO system, and in-house purified, reconstituted enzyme) to determine whether the enzyme systems exhibited kinetic differences in metabolic product formation rates under uniform experimental conditions. Warfarin 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 16753237-1 2006 Biotransformation of deoxypodophyllotoxin to epipodophyllotoxin by three major human hepatic enzymes, CYP1A2, CYP2C9 and CYP3A4, heterologously expressed in E. coli DH5alpha, was investigated. deoxypodophyllotoxin 21-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 16753237-1 2006 Biotransformation of deoxypodophyllotoxin to epipodophyllotoxin by three major human hepatic enzymes, CYP1A2, CYP2C9 and CYP3A4, heterologously expressed in E. coli DH5alpha, was investigated. Podophyllotoxin 45-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 17085674-2 2006 Functional polymorphisms of UGT1A6 (T181A and R184S) and CYP2C9 (R144C and I359L) have been reported to modify the protective effect of aspirin on colorectal adenoma risk. Aspirin 136-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16784736-13 2006 CONCLUSIONS: The BCRP 421C>A polymorphism may play an important role in the pharmacokinetics of rosuvastatin in healthy Chinese males after the exclusion of impact of SLCO1B1 and CYP2C9 genetic polymorphism. Rosuvastatin Calcium 99-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 17112811-0 2006 Impact of CYP2C9*3/*3 genotype on the pharmacokinetics and pharmacodynamics of piroxicam. Piroxicam 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 17112813-0 2006 Warfarin dose requirement for patients with both VKORC1 3673A/A and CYP2C9*3/*3 genotypes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 16928789-7 2006 In addition, when (S)-flurbiprofen was used as a substrate probe to determine CYP2C9 inhibition with a set of 12 inhibitors, decreased inhibition potency was observed across 11 of those inhibitors in the RECO purified, reconstituted enzyme compared with the Supersomes baculovirus microsomal preparation and pooled human liver microsomes. Flurbiprofen 18-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 16869821-4 2006 Results The acenocoumarol-sensitive patient was found to possess, in addition to CYP2C9*3 allele, a CYP2C9*11 allele and the VKORC1 AA diplotype which were all traced back through the parental lines. Acenocoumarol 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 16869821-5 2006 Conclusions Acenocoumarol sensitivity in this subject is the consequence of inheritance of multiple functionally defective alleles in both the CYP2C9 and VKORC1 genes. Acenocoumarol 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 17015052-0 2006 A warfarin-dosing model in Asians that uses single-nucleotide polymorphisms in vitamin K epoxide reductase complex and cytochrome P450 2C9. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-138 17064211-9 2006 Warfarin undergoes hepatic metabolism through cytochrome P450 2C9, and carbamazepine induces this isoenzyme. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 16740353-4 2006 CYP2C9*13, firstly identified by some of the present authors in a Chinese poor metabolizer of lornoxicam, is characterized by mutation encoding Leu90Pro substitution. lornoxicam 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16740353-4 2006 CYP2C9*13, firstly identified by some of the present authors in a Chinese poor metabolizer of lornoxicam, is characterized by mutation encoding Leu90Pro substitution. leu90pro 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16740353-5 2006 Kinetic experiments show that CYP2C9*13 has less catalytic activity in elimination of diclofenac and lornoxicam in vitro. Diclofenac 86-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 16740353-5 2006 Kinetic experiments show that CYP2C9*13 has less catalytic activity in elimination of diclofenac and lornoxicam in vitro. lornoxicam 101-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 16740353-7 2006 The structure change caused by Leu90Pro replacement is revealed and used to explain the dramatic decrease of the enzymatic activity in clearance of the two CYP2C9 substrates: diclofenac and lornoxicam. leu90pro 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 16740353-7 2006 The structure change caused by Leu90Pro replacement is revealed and used to explain the dramatic decrease of the enzymatic activity in clearance of the two CYP2C9 substrates: diclofenac and lornoxicam. Diclofenac 175-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 16740353-7 2006 The structure change caused by Leu90Pro replacement is revealed and used to explain the dramatic decrease of the enzymatic activity in clearance of the two CYP2C9 substrates: diclofenac and lornoxicam. lornoxicam 190-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 16815961-0 2006 Amiodarone analog-dependent effects on CYP2C9-mediated metabolism and kinetic profiles. Amiodarone 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 17015052-1 2006 INTRODUCTION: Because of the unique lack of genetic diversity despite the multiethnicity in the Asian population, we hypothesize that single-nucleotide polymorphisms in cytochrome P450 (CYP) 2C9 (CYP2C9*3) and vitamin K epoxide reductase complex subunit 1 (VKORC1) at position 381, used to infer VKORC1haplotype in combination with demographic factors, can accurately predict warfarin doses. Warfarin 376-384 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-194 16815961-5 2006 Amiodarone activated naproxen demethylation at lower concentrations, regardless of the CYP2C9 allele, and inhibited metabolism at higher concentrations without altering the kinetic profile. Amiodarone 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 17015052-1 2006 INTRODUCTION: Because of the unique lack of genetic diversity despite the multiethnicity in the Asian population, we hypothesize that single-nucleotide polymorphisms in cytochrome P450 (CYP) 2C9 (CYP2C9*3) and vitamin K epoxide reductase complex subunit 1 (VKORC1) at position 381, used to infer VKORC1haplotype in combination with demographic factors, can accurately predict warfarin doses. Warfarin 376-384 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 16815961-7 2006 Benzbromarone altered naproxen demethylation kinetics from a biphasic profile to that of a hyperbolic form in CYP2C9.1 and CYP2C9.3, resulting in inhibition and activation, respectively. Benzbromarone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 16815961-7 2006 Benzbromarone altered naproxen demethylation kinetics from a biphasic profile to that of a hyperbolic form in CYP2C9.1 and CYP2C9.3, resulting in inhibition and activation, respectively. Benzbromarone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 16815961-7 2006 Benzbromarone altered naproxen demethylation kinetics from a biphasic profile to that of a hyperbolic form in CYP2C9.1 and CYP2C9.3, resulting in inhibition and activation, respectively. Naproxen 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 17015052-8 2006 CONCLUSION: Warfarin dose requirements in Asians can be accurately predicted by use of a combination of patient demographics and a simplified genotyping approach for single variants in CYP2C9 and VKORC1. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. Sulfur 46-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16815961-8 2006 In contrast, benz(meth)arone activated naproxen demethylation in both CYP2C9.1 and CYP2C9.3. benz(meth)arone 13-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 16815961-8 2006 In contrast, benz(meth)arone activated naproxen demethylation in both CYP2C9.1 and CYP2C9.3. benz(meth)arone 13-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 16815961-8 2006 In contrast, benz(meth)arone activated naproxen demethylation in both CYP2C9.1 and CYP2C9.3. Naproxen 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 16815961-8 2006 In contrast, benz(meth)arone activated naproxen demethylation in both CYP2C9.1 and CYP2C9.3. Naproxen 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 16862439-2 2006 Presented here are the results on the construction and characterization of three fusion proteins containing the N-terminally modified human cytochrome P450s CYP2C9, CY2C19 and CYP3A4 fused to the soluble NADPH-dependent oxidoreductase domain of CYP102A1 from Bacillus megaterium. NADP 204-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 16946524-2 2006 S-Warfarin 7-hydroxylation by recombinant CYP2C9.1 and CYP2C9.3 was inhibited by glimepiride competitively. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 16714062-6 2006 Among the statins, simvastatin, lovastatin and atorvastatin are metabolized by cytochrome P450 3A4 (CYP3A4) while fluvastatin is metabolized by CYP2C9. Atorvastatin 47-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 16714062-6 2006 Among the statins, simvastatin, lovastatin and atorvastatin are metabolized by cytochrome P450 3A4 (CYP3A4) while fluvastatin is metabolized by CYP2C9. Fluvastatin 114-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 16946524-2 2006 S-Warfarin 7-hydroxylation by recombinant CYP2C9.1 and CYP2C9.3 was inhibited by glimepiride competitively. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. Sulfur 46-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16946524-2 2006 S-Warfarin 7-hydroxylation by recombinant CYP2C9.1 and CYP2C9.3 was inhibited by glimepiride competitively. glimepiride 81-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16946524-2 2006 S-Warfarin 7-hydroxylation by recombinant CYP2C9.1 and CYP2C9.3 was inhibited by glimepiride competitively. glimepiride 81-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 16946524-3 2006 The apparent K(i) value of glimepiride was lower at CYP2C9.3 than at CYP2C9.1. glimepiride 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16946524-3 2006 The apparent K(i) value of glimepiride was lower at CYP2C9.3 than at CYP2C9.1. glimepiride 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 16952492-1 2006 BACKGROUND: We have previously shown that flurbiprofen metabolism to 4"-hydroxyflurbiprofen provides an in vivo measure of cytochrome P450 (CYP) 2C9 activity. Flurbiprofen 42-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-148 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16946524-4 2006 Glimepiride also inhibited 7-hydroxylation of S-warfarin in a competitive manner by microsomes from human liver which showed the genotypes of CYP2C9, as CYP2C9*1/*1 or CYP2C9*1/*3. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 16940139-0 2006 Safety of voriconazole in a patient with CYP2C9*2/CYP2C9*2 genotype. Voriconazole 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 16940139-0 2006 Safety of voriconazole in a patient with CYP2C9*2/CYP2C9*2 genotype. Voriconazole 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 16952496-0 2006 Is celecoxib an inducer of cytochrome P450 3A4 in subjects carrying the CYP2C9*3 allele? Celecoxib 3-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 16952492-1 2006 BACKGROUND: We have previously shown that flurbiprofen metabolism to 4"-hydroxyflurbiprofen provides an in vivo measure of cytochrome P450 (CYP) 2C9 activity. 4'-hydroxyflurbiprofen 69-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-148 16847664-2 2006 The CYP2C9 genotype has been shown to influence the response to warfarin therapy, but such an effect on phenprocoumon therapy remains uncertain. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 16763015-7 2006 Hepatocytes exposed to 1 mM FFA for 14 h showed lower activity values of CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 enzymes than nontreated hepatocytes (about 45-65% reduction). Fatty Acids, Nonesterified 28-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 16815679-3 2006 CYP2C9 catalyses the prochiral formation of (R) and (S)-p-HPPH, and is approximately 40 times more stereoselective towards the formation of the (S) isomer whereas CYP2C19 is not stereoselective. Ranolazine 44-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16815679-3 2006 CYP2C9 catalyses the prochiral formation of (R) and (S)-p-HPPH, and is approximately 40 times more stereoselective towards the formation of the (S) isomer whereas CYP2C19 is not stereoselective. (s)-p-hpph 52-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16888441-4 2006 RECENT FINDINGS: Single-nucleotide polymorphisms in cytochrome P450 2C9 and vitamin K epoxide reductase have been shown to make significant contributions to the variability in warfarin dose requirements. Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 16611750-3 2006 OBJECTIVE: We tested for association between single nucleotide polymorphisms (SNPs) in VKORC1 and CYP2C9 and average weekly warfarin dose required to maintain patients at their desired anticoagulation target. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 16611750-10 2006 CYP2C9*3 was significantly (p = 0.05) associated with average warfarin dosage after adjustment for VKORC1*1173. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16611750-12 2006 The consistent findings regarding the role of VKORC1 and CYP2C9 in warfarin metabolism and maintenance dosage represent a clinically useful proof of principal for the use of pharmacogenomic information in medicine and may lead to improved understanding of warfarin"s actions. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16918308-4 2006 Potential mechanisms for oxazaphosphorine resistance include decreased activation by cytochrome P450s (e.g. CYP3A4, CYP2C9 and CYP2B6), increased deactivation of the agents by deactivating enzymes such as aldehyde dehydrogenases (ALDHs), increased cellular thiol level, increased DNA repair capacity, and altered cellular apoptotic response to DNA repair, e.g. deficient apoptosis due to lack of cellular mechanisms to result in cell death following DNA damage. oxazaphosphorine 25-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 16890578-10 2006 The mean daily dose of warfarin was lower in subjects with CYP2C9*1/*3 (1.86 +/- 0.80 mg/d, P = .007) than in subjects with CYP2C9*1/*1 (3.36 +/- 1.43 mg/d). Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 16890578-10 2006 The mean daily dose of warfarin was lower in subjects with CYP2C9*1/*3 (1.86 +/- 0.80 mg/d, P = .007) than in subjects with CYP2C9*1/*1 (3.36 +/- 1.43 mg/d). Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 16679385-4 2006 Using a substrate-cassette approach, P450 activities were determined after incubation with the prototypic inhibitors tienilic acid (CYP2C9), erythromycin, troleandomycin, and fluoxetine (CYP3A4). Ticrynafen 117-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 16679385-6 2006 Tienilic acid selectively inhibited CYP2C9-dependent diclofenac 4"-hydroxylation activity, and erythromycin, troleandomycin, and fluoxetine inhibited CYP3A4-dependent midazolam 1"-hydroxylation in a time- and concentration-dependent manner. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 16679385-6 2006 Tienilic acid selectively inhibited CYP2C9-dependent diclofenac 4"-hydroxylation activity, and erythromycin, troleandomycin, and fluoxetine inhibited CYP3A4-dependent midazolam 1"-hydroxylation in a time- and concentration-dependent manner. Diclofenac 53-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 16580879-1 2006 Polymorphisms in the CYP2C9 gene can have a significant impact on drug therapy by affecting the pharmacokinetics of frequently prescribed drugs, such as phenprocoumon and warfarin. Phenprocoumon 153-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 16698161-6 2006 Sulphaphenazole, a specific human CYP2C9 inhibitor, showed a Ki value of 30.8 microM and IC50 at 44.0 microM in the test system. Sulfaphenazole 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 16580879-1 2006 Polymorphisms in the CYP2C9 gene can have a significant impact on drug therapy by affecting the pharmacokinetics of frequently prescribed drugs, such as phenprocoumon and warfarin. Warfarin 171-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. endothelium-derived hyperpolarizing factor 74-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-55 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. endothelium-derived hyperpolarizing factor 74-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-64 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. edhf 118-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-55 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. edhf 118-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-64 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-55 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-64 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. Sulfaphenazole 225-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-55 16875977-1 2006 OBJECTIVES: We assessed the role of cytochrome P450 2C9 (CYP 2C9)-derived endothelium-derived hyperpolarizing factor (EDHF) in the forearm microcirculation of essential hypertensive patients (EH) by utilizing sulfaphenazole (SUL), a selective CYP 2C9 inhibitor. Sulfaphenazole 225-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-64 16875977-3 2006 Cytochrome P450 2C9 is a possible source of EDHF. edhf 44-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 17024799-0 2006 Tamoxifen inhibits cytochrome P450 2C9 activity in breast cancer patients. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-38 17024799-1 2006 Tamoxifen has been reported to potentiate the anticoagulant effect of warfarin and also to increase the plasma level of phenytoin, which are mainly metabolized by CYP2C9. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 17024799-1 2006 Tamoxifen has been reported to potentiate the anticoagulant effect of warfarin and also to increase the plasma level of phenytoin, which are mainly metabolized by CYP2C9. Phenytoin 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 17024799-2 2006 The aim of this study was to determine the influence of tamoxifen on CYP2C9 activity in vivo in humans. Tamoxifen 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 17024799-7 2006 Tamoxifen inhibited CYP2C9 activity in breast cancer patients within two weeks of its administration. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 17024799-8 2006 The inhibition of CYP2C9 activity may be a possible explanation for the drug-drug interaction of tamoxifen with CYP2C9 substrates. Tamoxifen 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 17024799-8 2006 The inhibition of CYP2C9 activity may be a possible explanation for the drug-drug interaction of tamoxifen with CYP2C9 substrates. Tamoxifen 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 29788657-5 2006 Thus, cytochrome P450 (CYP)2D6 polymorphism influences codeine and tramadol analgesic effects, CYP2C9 has an impact on the disposition of some nonsteroidal anti-inflammatory drugs, and opioid receptor polymorphism (118A>G) may reduce morphine potency. Morphine 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 16960144-3 2006 It is now clear that genetic polymorphisms in genes influencing metabolism (CYP2C9) and pharmacodynamic response (VKORC1) are strongly associated with warfarin responsiveness. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 16847429-6 2006 Multivariate regression analysis showed that patient age, height and CYP2C9, VKORC1 and APOE genotypes significantly contributed to warfarin dose requirement (R = 57%). Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 16802783-4 2006 Here, we have attached CYP2C9 to gold substrates such that the resulting construct maintains the ability to bind and metabolize substrates in the presence of NADPH and cytochrome P450 reductase. NADP 158-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 16867169-4 2006 In detail these selective substrates were caffeine (CYP1A2), coumarin (CYP2A6), tolbutamide (CYP2C9), S-(+)-mephenytoin (CYP2C19), dextromethorphane (CYP2D6), chlorzoxazone (CYP2E1) and testosterone (CYP3A4). Tolbutamide 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 16867170-3 2006 The metabolism of pioglitazone by CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4 and CYP3A5 was investigated using human recombinant CYP isoforms. Pioglitazone 18-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 16818632-6 2006 In NHFs cultured in low versus normal serum, MYC induced increased expression of CYP2C9, a gene product well known to be associated with ROS production. Reactive Oxygen Species 137-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 16818632-7 2006 Specific inhibition of CYP2C9 by small interfering RNA was shown to partially inhibit MYC-induced ROS production. Reactive Oxygen Species 98-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 16797247-5 2006 RESULTS: Variant CYP2C9 genotypes enhanced the protective effect of ibuprofen on the prevention of colorectal cancer, and a dose-response relationship with respect to increasing numbers of variant alleles was seen (P interaction = .02). Ibuprofen 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 16797247-8 2006 CONCLUSIONS: In this study, the major effect seen was an enhancement by slower-metabolizing CYP2C9 variants of the chemopreventive activity of ibuprofen against colorectal cancer. Ibuprofen 143-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 16783563-0 2006 Amodiaquine, its desethylated metabolite, or both, inhibit the metabolism of debrisoquine (CYP2D6) and losartan (CYP2C9) in vivo. Amodiaquine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16783563-0 2006 Amodiaquine, its desethylated metabolite, or both, inhibit the metabolism of debrisoquine (CYP2D6) and losartan (CYP2C9) in vivo. Debrisoquin 77-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16783563-0 2006 Amodiaquine, its desethylated metabolite, or both, inhibit the metabolism of debrisoquine (CYP2D6) and losartan (CYP2C9) in vivo. Losartan 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16783563-1 2006 OBJECTIVE: To study the extent of in vivo inhibition by the antimalarial drug amodiaquine, its active metabolite N-desethylamodiaquine, or both, of the metabolism of four probe drugs of the enzymes CYP2D6, CYP2C19, CYP2C9 and CYP1A2. Amodiaquine 78-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 215-221 16783563-8 2006 The CYP2D6 and CYP2C9 activities of the subjects were measured by debrisoquine and losartan phenotyping tests, respectively. Debrisoquin 66-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16783563-8 2006 The CYP2D6 and CYP2C9 activities of the subjects were measured by debrisoquine and losartan phenotyping tests, respectively. Losartan 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16783563-10 2006 The effects on CYP2D6 and CYP2C9 activities subsided within a week after intake of amodiaquine as tested by the phenotyping cocktail. Amodiaquine 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 16783563-12 2006 CONCLUSION: A single dose of amodiaquine decreased CYP2D6 and CYP2C9 activities significantly compared to baseline values. Amodiaquine 29-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 16723553-11 2006 This was likely caused by inhibition of the cytochrome P450 2C9-mediated metabolism of S-(+)-ibuprofen. Ibuprofen 87-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-63 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. aceclofenac 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Celecoxib 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Diclofenac 130-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Ibuprofen 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Indomethacin 153-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. lornoxicam 167-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Piroxicam 179-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 17022718-11 2006 The risk for digestive hemorrhage associated with the CYP2C9 genotype is particularly relevant when using aceclofenac, celecoxib, diclofenac, ibuprofen, indomethacin, lornoxicam, piroxicam, or naproxen. Naproxen 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 16788382-1 2006 Genetic variations in cytochrome P450 2C9 (CYP2C9) are known to contribute to interindividual and interethnic variability in response to clinical drugs such as warfarin. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-41 16788382-1 2006 Genetic variations in cytochrome P450 2C9 (CYP2C9) are known to contribute to interindividual and interethnic variability in response to clinical drugs such as warfarin. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 16639745-0 2006 Multiple molecular dynamics simulations of human p450 monooxygenase CYP2C9: the molecular basis of substrate binding and regioselectivity toward warfarin. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 16639745-4 2006 Besides the nonproductive state of the CYP2C9 warfarin complex captured in the crystal structure, three additional states were observed. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 16722840-7 2006 More recently, single nucleotide polymorphisms in the 2C9 isoform of cytochrome P450 (CYP2C9) and vitamin K epoxide reductase (VKOR) have been shown to make significant contributions to the variability in coumarin dosage requirements. coumarin 205-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 16630605-0 2006 Frequency of CYP2C9 polymorphisms affecting warfarin metabolism in a large anticoagulant clinic cohort. Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16630605-7 2006 CONCLUSIONS: Allele frequencies and genotypes for CYP2C9*2 and *3 variants in patients on warfarin are not statistically different from controls whether or not they are stratified for ethnicity. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 16630605-9 2006 CYP2C9 genotype predicts warfarin dosage even in an uncontrolled, retrospective survey of unselected patients on warfarin therapy. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16630605-9 2006 CYP2C9 genotype predicts warfarin dosage even in an uncontrolled, retrospective survey of unselected patients on warfarin therapy. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16501008-9 2006 The K(i) values for CYP2C9 and CYP2C19 were 1.7 and 3.3 microM, respectively, and those for CYP3A4 were 8.3 and 2.9 microM, using two substrates, testosterone and midazolam, respectively. Testosterone 146-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 16325295-0 2006 Effect of CYP2C9 genetic polymorphisms on the efficacy and pharmacokinetics of glimepiride in subjects with type 2 diabetes. glimepiride 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 16325295-1 2006 Glimepiride, a sulfonylurea hypoglycemic agent, is metabolized by cytochrome P450 2C9 (CYP2C9) which is known to have genetic polymorphisms. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-85 16325295-1 2006 Glimepiride, a sulfonylurea hypoglycemic agent, is metabolized by cytochrome P450 2C9 (CYP2C9) which is known to have genetic polymorphisms. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 16325295-4 2006 The long-term observations of 2 patients with a CYP2C9*1/*3 suggested that subjects with a CYP2C9*1/*3 respond well to glimepiride during the initial phase of treatment, but 1 patient have shown the weight gain over the long-term treatment. glimepiride 119-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 16325295-4 2006 The long-term observations of 2 patients with a CYP2C9*1/*3 suggested that subjects with a CYP2C9*1/*3 respond well to glimepiride during the initial phase of treatment, but 1 patient have shown the weight gain over the long-term treatment. glimepiride 119-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 16325295-5 2006 The pharmacokinetic study showed that the area under the concentration-time curve for glimepiride in the CYP2C9*1/*3 subjects was approximately 2.5-fold higher than that of the CYP2C9*1/*1 subjects. glimepiride 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 16325295-5 2006 The pharmacokinetic study showed that the area under the concentration-time curve for glimepiride in the CYP2C9*1/*3 subjects was approximately 2.5-fold higher than that of the CYP2C9*1/*1 subjects. glimepiride 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 16325295-6 2006 The intrinsic clearance of glimepiride by the CYP2C9*3 enzyme was lower than that by the CYP2C9*1 enzyme. glimepiride 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16325295-6 2006 The intrinsic clearance of glimepiride by the CYP2C9*3 enzyme was lower than that by the CYP2C9*1 enzyme. glimepiride 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 16325295-7 2006 These results suggested that the lower hydroxylation activity of glimepiride in the subject with type 2 diabetes and CYP2C9*1/*3 led to a marked elevation in the plasma concentrations of glimepiride and a stronger pharmacological effect of glimepiride. glimepiride 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 16325295-7 2006 These results suggested that the lower hydroxylation activity of glimepiride in the subject with type 2 diabetes and CYP2C9*1/*3 led to a marked elevation in the plasma concentrations of glimepiride and a stronger pharmacological effect of glimepiride. glimepiride 187-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 16325295-7 2006 These results suggested that the lower hydroxylation activity of glimepiride in the subject with type 2 diabetes and CYP2C9*1/*3 led to a marked elevation in the plasma concentrations of glimepiride and a stronger pharmacological effect of glimepiride. glimepiride 187-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Lopinavir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 16639344-0 2006 Lopinavir/ritonavir induces the hepatic activity of cytochrome P450 enzymes CYP2C9, CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal activity of CYP3A as measured by a phenotyping drug cocktail in healthy volunteers. Ritonavir 10-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 16676068-1 2006 The degree of interpatient variability in the warfarin dose required to achieve the desired anticoagulant response can only partly be explained by polymorphisms in the CYP2C9 gene, suggesting that additional genetic factors such as polymorphisms in genes involved in blood coagulation may influence warfarin dose requirement. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 16676068-1 2006 The degree of interpatient variability in the warfarin dose required to achieve the desired anticoagulant response can only partly be explained by polymorphisms in the CYP2C9 gene, suggesting that additional genetic factors such as polymorphisms in genes involved in blood coagulation may influence warfarin dose requirement. Warfarin 299-307 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 16676068-8 2006 With a regression model we explained 60.0% of variability in warfarin dose, which was due to gene polymorphisms (CYP2C9, VKORC1), age and body-surface-area. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16627267-2 2006 We developed and verified a melting analysis assay for common single nucleotide polymorphisms of cytochrome P-450 (CYP) 2C9 that affect warfarin metabolism. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-123 16580898-0 2006 Influence of coagulation factor, vitamin K epoxide reductase complex subunit 1, and cytochrome P450 2C9 gene polymorphisms on warfarin dose requirements. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 16580898-1 2006 INTRODUCTION: The primary objective of this study was to determine whether variability in warfarin dose requirements is determined by common polymorphisms in genes whose products are involved in the pharmacodynamics and pharmacokinetics of warfarin, namely, the coagulation factors, vitamin K epoxide reductase complex subunit 1 (VKORC1), and cytochrome P450 (CYP) 2C9. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 343-368 16580898-1 2006 INTRODUCTION: The primary objective of this study was to determine whether variability in warfarin dose requirements is determined by common polymorphisms in genes whose products are involved in the pharmacodynamics and pharmacokinetics of warfarin, namely, the coagulation factors, vitamin K epoxide reductase complex subunit 1 (VKORC1), and cytochrome P450 (CYP) 2C9. Warfarin 240-248 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 343-368 16552506-2 2006 We have previously shown that warfarin dose is influenced by cytochrome P450 (CYP) 2C9 genotype, age, body weight and co-treatment with drugs that interfere with warfarin metabolism. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-86 16552506-2 2006 We have previously shown that warfarin dose is influenced by cytochrome P450 (CYP) 2C9 genotype, age, body weight and co-treatment with drugs that interfere with warfarin metabolism. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-86 16556082-7 2006 The systemic azoles, such as ketoconazole, itraconazole, fluconazole and voriconazole, are inhibitors of CYP isoenzymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Azoles 13-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 16556082-7 2006 The systemic azoles, such as ketoconazole, itraconazole, fluconazole and voriconazole, are inhibitors of CYP isoenzymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Ketoconazole 29-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 16556082-7 2006 The systemic azoles, such as ketoconazole, itraconazole, fluconazole and voriconazole, are inhibitors of CYP isoenzymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Itraconazole 43-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 16556082-7 2006 The systemic azoles, such as ketoconazole, itraconazole, fluconazole and voriconazole, are inhibitors of CYP isoenzymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Fluconazole 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 16556082-7 2006 The systemic azoles, such as ketoconazole, itraconazole, fluconazole and voriconazole, are inhibitors of CYP isoenzymes, such as CYP3A4, CYP2C9 and CYP2C19, to varying degrees. Voriconazole 73-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 16505204-5 2006 Aerosol gene transfer of recombinant adenovirus containing the human CYP2C9 significantly elevated mean pulmonary artery pressure and total pulmonary resistance indices, both of which were sensitive to the inhibitor sulfaphenazole. Sulfaphenazole 216-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 16765138-0 2006 CYP2C9 genotyping in acenocoumarol treatment: is it a cost-effective addition to international normalized ratio monitoring? Acenocoumarol 21-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16793587-4 2006 CYP2C9, for instance, was involved in the conversion of estrogen sulfate to 16-hydroxy sulfate metabolite, DDC in estrogen-dependent pathogenesis of endometrial carcinoma possibly by DDC interaction with AR to enhance steroid receptor transcription. estrogen sulfate 56-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16793587-4 2006 CYP2C9, for instance, was involved in the conversion of estrogen sulfate to 16-hydroxy sulfate metabolite, DDC in estrogen-dependent pathogenesis of endometrial carcinoma possibly by DDC interaction with AR to enhance steroid receptor transcription. 16-hydroxy sulfate 76-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16793587-4 2006 CYP2C9, for instance, was involved in the conversion of estrogen sulfate to 16-hydroxy sulfate metabolite, DDC in estrogen-dependent pathogenesis of endometrial carcinoma possibly by DDC interaction with AR to enhance steroid receptor transcription. Zalcitabine 107-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16793587-4 2006 CYP2C9, for instance, was involved in the conversion of estrogen sulfate to 16-hydroxy sulfate metabolite, DDC in estrogen-dependent pathogenesis of endometrial carcinoma possibly by DDC interaction with AR to enhance steroid receptor transcription. Zalcitabine 183-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16547074-5 2006 It is noteworthy that Compound K, protopanaxadiol (Ppd), and protopanaxatriol (Ppt) all exhibited moderate inhibition against CYP2C9 activity, and Ppd and Ppt also exhibited potent competitive inhibition against CYP3A4 activity. protopanaxadiol 34-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 16547074-5 2006 It is noteworthy that Compound K, protopanaxadiol (Ppd), and protopanaxatriol (Ppt) all exhibited moderate inhibition against CYP2C9 activity, and Ppd and Ppt also exhibited potent competitive inhibition against CYP3A4 activity. protopanaxatriol 61-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 16547074-5 2006 It is noteworthy that Compound K, protopanaxadiol (Ppd), and protopanaxatriol (Ppt) all exhibited moderate inhibition against CYP2C9 activity, and Ppd and Ppt also exhibited potent competitive inhibition against CYP3A4 activity. protopanaxatriol 79-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 16413010-2 2006 Cytochrome P4502C9 (CYP2C9) is polymorphic in human and is principally responsible for the metabolism of warfarin. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-18 16413010-2 2006 Cytochrome P4502C9 (CYP2C9) is polymorphic in human and is principally responsible for the metabolism of warfarin. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 16413010-4 2006 We screened a new polymorphism of CYP2C9 and investigated its role in warfarin sensitivity. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 16413010-10 2006 The association between warfarin dose requirement and genetic polymorphism of CYP2C9 was also analysed. Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 16501008-8 2006 Using cDNA-expressed enzymes, clevidipine inhibited CYP2C9, CYP2C19, and CYP3A4 activities with IC(50) values below 10 microM, whereas CYP1A2, CYP2D6, and CYP2E1 activities were not substantially inhibited (IC(50) values >70 microM). clevidipine 30-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 16873909-0 2006 Influence of the CYP2C9 AND CYP2C19 polymorphisms on phenytoin hydroxylation in healthy individuals from south India. Phenytoin 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 16873909-1 2006 BACKGROUND AND OBJECTIVES: Phenytoin, a widely used anti-epileptic drug, is metabolized mainly by CYP2C9 (90%) and partly by CYP2C19 (10%) to its major metabolite 5-(para-hydroxyphenyl)-5- phenylhydantoin (p-HPPH). Phenytoin 27-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 16873909-8 2006 RESULTS: A significant correlation was observed between the CYP2C9 genotype and metabolic ratio of phenytoin/p-HPPH (r = 0.472, 95% CI 0.100 to 0.728; P = 0.01). Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 16873909-9 2006 While no association was found with CYP2C19 alone, a significant correlation was observed between the combined CYP2C9 and CYP2C19 genotypes and phenytoin metabolic ratio (r = 0.507, 95% CI 0.146 to 0.749; P< 0.01). Phenytoin 144-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 16873909-10 2006 INTERPRETATION AND CONCLUSION: CYP2C9*2 and *3 mutant alleles caused decreased hydroxylation of phenytoin in vivo, whereas the mutant alleles of CYP2C19 played only a minor role in the metabolism of phenytoin in subjects of our study. Phenytoin 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 16873909-10 2006 INTERPRETATION AND CONCLUSION: CYP2C9*2 and *3 mutant alleles caused decreased hydroxylation of phenytoin in vivo, whereas the mutant alleles of CYP2C19 played only a minor role in the metabolism of phenytoin in subjects of our study. Phenytoin 199-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 16639344-6 2006 CYP2C9 and CYP2C19 activities were quantified by S-WARF AUC0-inf and OMP/5-hydroxy OMP ratio, respectively. 5-hydroxymethylomeprazole 73-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16318905-6 2006 CYP1A2 and CYP2C9 played minor roles in the metabolism of rutaecarpine. rutecarpine 58-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 16708125-2 2006 One of several factors underlying the variability in warfarin dose response in the patients receiving this oral anticoagulant is a genetic predisposition, especially the CYP2C9 polymorphisms. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 16708125-7 2006 The clinical correlation between CYP2C9 polymorphism and warfarin metabolism was also assessed. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 16708125-9 2006 CYP2C9 variants are strongly associated with low-dose warfarin requirement. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16708125-10 2006 In summary, the CYP2C9 variants strongly affect the warfarin dose requirement. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 16415119-7 2006 A monoclonal antibody to CYP2B6 and the CYP3A inhibitor ketoconazole substantially inhibited R-138727 formation, whereas inhibitors of CYP2C9 (sulfaphenazole) and CYP2C19 (omeprazole) did not. Sulfaphenazole 143-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 16501008-9 2006 The K(i) values for CYP2C9 and CYP2C19 were 1.7 and 3.3 microM, respectively, and those for CYP3A4 were 8.3 and 2.9 microM, using two substrates, testosterone and midazolam, respectively. Midazolam 163-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 16699986-3 2006 AIM: To investigate the influence of CYP2C9 polymorphism on the occurrence of bleeding complications related to acenocoumarol therapy. Acenocoumarol 112-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 16635054-1 2006 BACKGROUND: CYP2C9 is one of the major drug metabolizing enzymes for many drugs including warfarin, NSAIDs and losartan. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16635054-1 2006 BACKGROUND: CYP2C9 is one of the major drug metabolizing enzymes for many drugs including warfarin, NSAIDs and losartan. Losartan 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16513453-0 2006 Accumulation of celecoxib with a 7-fold higher drug exposure in individuals homozygous for CYP2C9*3. Celecoxib 16-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 16490777-1 2006 7-Methoxy-4-trifluoromethylcoumarin (MFC) has been used extensively in high-throughput screens for the identification of potential CYP2C9 interactions. 7-methoxy-4-trifluoromethylcoumarin 0-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 16490777-1 2006 7-Methoxy-4-trifluoromethylcoumarin (MFC) has been used extensively in high-throughput screens for the identification of potential CYP2C9 interactions. 7-methoxy-4-trifluoromethylcoumarin 37-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 16513444-5 2006 CYP2C9*2 and VKORC1 coding region variants were rare (<2%), whereas CYP2C9*3 associated with lower warfarin requirements was less common in Chinese and Malays (7% and 9%, respectively) than in Indians (18%) and could not account for their lower warfarin requirements. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16699986-6 2006 RESULTS: In 145 patients bearing alleles with reduced activity (CYP2C9*2 and/or *3), the optimal dose of acenocoumarol was significantly (p<0.001) lower than in patients with the wild type allele (2.12+/-0.96 mg/day and 2.90+/-1.46 mg/day, respectively). Acenocoumarol 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 16699986-7 2006 In comparison with wild type allele patients, the mean daily acenocoumarol dose was lower in the CYP2C9*2 group, and the lowest in *3 bearers. Acenocoumarol 61-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 16699986-9 2006 CONCLUSIONS: Patients bearing CYP2C9 alleles with reduced enzymatic activity have a lower acenocoumarol requirement. Acenocoumarol 90-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 16445595-0 2006 Inhibitory effect of 5-fluorouracil on cytochrome P450 2C9 activity in cancer patients. Fluorouracil 21-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 16388406-5 2006 RESULTS: The acid forms had minimal inhibitory effects on all CYP activities tested, except for fluvastatin on CYP2C9-mediated tolbutamide 4-hydroxylation (IC50 = 1.7 microM) and simvastatin on CYP3A4/5-mediated paclitaxel 3-hydroxylation (12.0 microM). Fluvastatin 96-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 16388406-5 2006 RESULTS: The acid forms had minimal inhibitory effects on all CYP activities tested, except for fluvastatin on CYP2C9-mediated tolbutamide 4-hydroxylation (IC50 = 1.7 microM) and simvastatin on CYP3A4/5-mediated paclitaxel 3-hydroxylation (12.0 microM). Tolbutamide 127-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 16479536-6 2006 Tagging technology is shown to supersede conventional methods for P450 profiling in terms of discriminatory power and throughput, exemplified by the simultaneous detection of distinct induction profiles for cyp2c subfamily members in response to phenobarbitone: cyp2c29 expression, but not cyp2c40 or cyp2c50, was induced threefold by treatment. Phenobarbital 246-260 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-212 16445595-1 2006 Drug interactions have been reported between 5-fluorouracil and cytochrome P450 2C9 (CYP2C9) substrates, S-warfarin and phenytoin. Fluorouracil 45-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-83 16445595-1 2006 Drug interactions have been reported between 5-fluorouracil and cytochrome P450 2C9 (CYP2C9) substrates, S-warfarin and phenytoin. Fluorouracil 45-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 16445595-1 2006 Drug interactions have been reported between 5-fluorouracil and cytochrome P450 2C9 (CYP2C9) substrates, S-warfarin and phenytoin. Phenytoin 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 16445595-2 2006 This study was performed to determine the influence of 5-fluorouracil on cytochrome P450 2C9 (CYP2C9) activity in colorectal cancer patients (n=17) receiving 5-fluorouracil. Fluorouracil 55-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-92 16445595-2 2006 This study was performed to determine the influence of 5-fluorouracil on cytochrome P450 2C9 (CYP2C9) activity in colorectal cancer patients (n=17) receiving 5-fluorouracil. Fluorouracil 55-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 16445595-2 2006 This study was performed to determine the influence of 5-fluorouracil on cytochrome P450 2C9 (CYP2C9) activity in colorectal cancer patients (n=17) receiving 5-fluorouracil. Fluorouracil 158-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-92 16445595-2 2006 This study was performed to determine the influence of 5-fluorouracil on cytochrome P450 2C9 (CYP2C9) activity in colorectal cancer patients (n=17) receiving 5-fluorouracil. Fluorouracil 158-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 16445595-3 2006 Losartan was used as a marker to assess CYP2C9 activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 16445595-9 2006 The results suggest that in most patients 5-fluorouracil inhibited CYP2C9 activity. Fluorouracil 42-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 16445595-11 2006 This finding may help explain the mechanism of interaction between 5-fluorouracil and CYP2C9 substrates. Fluorouracil 67-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 16356485-3 2006 CI-1034 moderately inhibited CYP2C9 (IC(50) 39.6 microM) and CYP3A4 activity (IC(50) 21.6 microM); CYP3A4 inhibition was metabolism-dependent. PD 180988 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 16802848-10 2006 Clearance is hepatic via N-oxidation by the hepatic cytochrome P450 (CYP) isoenzymes, CYP2C19, CYP2C9 and CYP3A4. Nitrogen 25-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 16550733-1 2006 OBJECTIVE: Bosentan has been shown in vitro and in vivo to induce the cytochrome P450 enzymes CYP2C9 and CYP3A4. Bosentan 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 16863464-0 2006 Contribution of CYP2C9 to variability in vitamin K antagonist metabolism. Vitamin K 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 16863464-3 2006 CYP2C9 makes a very important contribution to metabolism of vitamin K antagonist anticoagulants, and is the main oxidising enzyme for S-warfarin and S-acenocoumarol as well as contributing to phenprocoumon metabolism. Vitamin K 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16863464-3 2006 CYP2C9 makes a very important contribution to metabolism of vitamin K antagonist anticoagulants, and is the main oxidising enzyme for S-warfarin and S-acenocoumarol as well as contributing to phenprocoumon metabolism. Warfarin 134-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16863464-3 2006 CYP2C9 makes a very important contribution to metabolism of vitamin K antagonist anticoagulants, and is the main oxidising enzyme for S-warfarin and S-acenocoumarol as well as contributing to phenprocoumon metabolism. Acenocoumarol 149-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16863464-4 2006 A large number of studies have now shown that CYP2C9 genotype predicts dose requirement for both warfarin and acenocoumarol, with a possible contribution for phenprocoumon. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16863464-4 2006 A large number of studies have now shown that CYP2C9 genotype predicts dose requirement for both warfarin and acenocoumarol, with a possible contribution for phenprocoumon. Acenocoumarol 110-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16863464-6 2006 Although CYP2C9 genotype is clearly a predictor of vitamin K antagonist dose requirement, especially in Caucasian populations in whom variant alleles are common, a number of recent studies have shown that age, genotype for the gene encoding the target gene vitamin K epoxide reductase and concomitant drugs are equally important factors in determining dose. Vitamin K 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 16424822-0 2006 Different contributions of polymorphisms in VKORC1 and CYP2C9 to intra- and inter-population differences in maintenance dose of warfarin in Japanese, Caucasians and African-Americans. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 16424822-10 2006 Furthermore, VKORC1 1173C > T and CYP2C9 (*2/*3/*11) genotypes, age and weight were identified as independent covariates contributing to interpatient variability in warfarin dosage. Warfarin 168-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 16424822-11 2006 CONCLUSIONS: Both VKORC1 and CYP2C9 polymorphisms contribute to inter-population difference in warfarin doses among the three populations, but their contribution to intra-population variability may differ within each population. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 16359930-12 2006 Currently, the best example of avoiding side effects is determining genotype at the CYP2C9 locus with respect of warfarin treatment, since carriers for functional variants (>20% of the population) require lower doses for optimal anticoagulation, and homozygotes, although rare, may well experience serious bleeding if given a usual dose. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 16493479-4 2006 Warfarin daily dose was predominantly determined by VKORC1 and CYP2C9 genotypes (partial r(2) = 0.21; 0.20, respectively). Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 16493479-7 2006 Combined genotype analysis revealed that CYP2C9 andVKORC1 wild type and CALU mutant patients required the highest warfarin doses (7.8 +/- 1.5mg/day; n = 9) as compared to the CYP2C9 and VKORC1 mutant and CALU wild type genotypes (2.8 +/- 0.3 mg/day; n = 18; p < 0.01). Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 16493479-9 2006 Compound genetic profiles comprising VKORC1, CALU and CYP2C9 improve categorization of individual warfarin dose requirements in more than 25% of patients at steady-state anticoagulation. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 16137701-2 2006 In this study the inhibitory effect of chamomile essential oil and its major constituents on four selected human cytochrome P450 enzymes (CYP1A2, CYP2C9, CYP2D6 and CYP3A4) was investigated. chamomile essential oil 39-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 16401082-6 2006 The oxidation of alpha-thujone by human CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 occurs with up to 80% C-4 methyl inversion, in agreement with a dominant radical hydroxylation mechanism. beta-thujone 17-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 16286479-1 2006 To determine the efficacy of cytochrome P450 2C9 metabolites of arachidonic acid, viz. Arachidonic Acid 64-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-48 16428919-3 2006 Therefore, in vitro and in vivo inhibition studies were done to ascertain the influence of ginkgo on CYP2C9, the P-450 isozyme responsible for the metabolism of the most potent warfarin enantiomer, (S)-warfarin. Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 16428919-3 2006 Therefore, in vitro and in vivo inhibition studies were done to ascertain the influence of ginkgo on CYP2C9, the P-450 isozyme responsible for the metabolism of the most potent warfarin enantiomer, (S)-warfarin. Warfarin 198-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 16475710-1 2006 The oxazaphosphorines cyclophosphamide (CP) and ifosfamide (IF) are alkylating agents that require bioactivation via cytochrome (CYP) P450 isoenzymes including CYP2C9 enzymes. oxazaphosphorines 4-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 16475710-1 2006 The oxazaphosphorines cyclophosphamide (CP) and ifosfamide (IF) are alkylating agents that require bioactivation via cytochrome (CYP) P450 isoenzymes including CYP2C9 enzymes. Cyclophosphamide 22-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 16475710-1 2006 The oxazaphosphorines cyclophosphamide (CP) and ifosfamide (IF) are alkylating agents that require bioactivation via cytochrome (CYP) P450 isoenzymes including CYP2C9 enzymes. Ifosfamide 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 16475710-3 2006 The mutant alleles of CYP2C9 were residue 144 (Arg (*1)/Cys (*2)), residue 358 (Tyr/Cys), residue 359 (Ile/Leu (*3)) and residue 417 (Gly/Asp). Arginine 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 16475710-3 2006 The mutant alleles of CYP2C9 were residue 144 (Arg (*1)/Cys (*2)), residue 358 (Tyr/Cys), residue 359 (Ile/Leu (*3)) and residue 417 (Gly/Asp). Cysteine 56-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 16475710-3 2006 The mutant alleles of CYP2C9 were residue 144 (Arg (*1)/Cys (*2)), residue 358 (Tyr/Cys), residue 359 (Ile/Leu (*3)) and residue 417 (Gly/Asp). Glycine 134-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 16475710-3 2006 The mutant alleles of CYP2C9 were residue 144 (Arg (*1)/Cys (*2)), residue 358 (Tyr/Cys), residue 359 (Ile/Leu (*3)) and residue 417 (Gly/Asp). Aspartic Acid 138-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 17112294-8 2006 Systemic clearance of dexloxiglumide is moderate and cytochrome P450 (CYP) 3A4/5 and CYP2C9 have been implicated in the metabolism of dexloxiglumide to produce O-demethyl dexloxi-glumide. dexloxiglumide 22-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17112294-8 2006 Systemic clearance of dexloxiglumide is moderate and cytochrome P450 (CYP) 3A4/5 and CYP2C9 have been implicated in the metabolism of dexloxiglumide to produce O-demethyl dexloxi-glumide. dexloxiglumide 134-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17112294-8 2006 Systemic clearance of dexloxiglumide is moderate and cytochrome P450 (CYP) 3A4/5 and CYP2C9 have been implicated in the metabolism of dexloxiglumide to produce O-demethyl dexloxi-glumide. o-demethyl dexloxi-glumide 160-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 17124101-3 2006 SNPs in the cytochrome P450 complex (CYP2C9) affect coumarin metabolism. coumarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 16413247-1 2006 OBJECTIVES: Recent anecdotal, unvalidated case reports have suggested potentiation of warfarin-induced anticoagulation by cranberry juice, possibly through inhibition of human cytochrome P450 (CYP) 2C9, the enzyme responsible for the clearance of the active S-enantiomer of warfarin. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-201 16413247-1 2006 OBJECTIVES: Recent anecdotal, unvalidated case reports have suggested potentiation of warfarin-induced anticoagulation by cranberry juice, possibly through inhibition of human cytochrome P450 (CYP) 2C9, the enzyme responsible for the clearance of the active S-enantiomer of warfarin. Warfarin 274-282 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-201 16299161-1 2006 Gemfibrozil more potently inhibits CYP2C9 than CYP2C8 in vitro, and yet the opposite inhibitory potency is observed in the clinic. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 16299161-3 2006 Gemfibrozil most potently inhibited CYP2C9 (IC50 of 30 microM), whereas gemfibrozil glucuronide most potently inhibited CYP2C8 (IC50 of 24 microM). Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 17124101-4 2006 Patients with either of two common variants, CYP2C9*2 or CYP2C9*3, metabolize coumarins slowly and are twice as likely to have a laboratory or clinical adverse event, unless their initial coumarin doses are reduced. Coumarins 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 17124101-4 2006 Patients with either of two common variants, CYP2C9*2 or CYP2C9*3, metabolize coumarins slowly and are twice as likely to have a laboratory or clinical adverse event, unless their initial coumarin doses are reduced. Coumarins 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 17124101-4 2006 Patients with either of two common variants, CYP2C9*2 or CYP2C9*3, metabolize coumarins slowly and are twice as likely to have a laboratory or clinical adverse event, unless their initial coumarin doses are reduced. coumarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 17124101-4 2006 Patients with either of two common variants, CYP2C9*2 or CYP2C9*3, metabolize coumarins slowly and are twice as likely to have a laboratory or clinical adverse event, unless their initial coumarin doses are reduced. coumarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16432637-0 2006 Association of VKORC1 and CYP2C9 polymorphisms with warfarin dose requirements in Japanese patients. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 16379042-2 2006 To assess the biotransformation and CYP reaction phenotype and their potential implications for human pharmacology and toxicology, the in vitro metabolism of ET-743 was characterized using incubations with human liver preparations, cytochrome P450 (CYP) and uridine diphosphoglucuronosyl transferase (UGT) supersomes.CYP supersomes and liver microsomes showed that ET-743 was metabolized mainly by CYP3A4, but also by CYP2C9, 2C19, 2D6, and 2E1. Trabectedin 158-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 418-424 16183265-3 2006 The pharmacokinetics of CPA and its active metabolite were related to the genotype of CYP2B6, CYP2C9 and CYP2C19. Cyclophosphamide 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 17827526-9 2006 It was located in the amino acid number 144 of CYP2C9 protein with the change of normal amino acid arginine into cysteine, which is the same as identified in poor metabolism patients as homozygous CYP2C9*2. amino acid arginine 88-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 17827526-9 2006 It was located in the amino acid number 144 of CYP2C9 protein with the change of normal amino acid arginine into cysteine, which is the same as identified in poor metabolism patients as homozygous CYP2C9*2. amino acid arginine 88-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 17827526-9 2006 It was located in the amino acid number 144 of CYP2C9 protein with the change of normal amino acid arginine into cysteine, which is the same as identified in poor metabolism patients as homozygous CYP2C9*2. Cysteine 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 17827526-9 2006 It was located in the amino acid number 144 of CYP2C9 protein with the change of normal amino acid arginine into cysteine, which is the same as identified in poor metabolism patients as homozygous CYP2C9*2. Cysteine 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 16432637-5 2006 We then genotyped the CYP2C9 gene for the Japanese common genetic variant, CYP2C9*3 and, based on the genotype of these two genes, classified patients into three categories, which we call "warfarin-responsive index." Warfarin 189-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 16432637-5 2006 We then genotyped the CYP2C9 gene for the Japanese common genetic variant, CYP2C9*3 and, based on the genotype of these two genes, classified patients into three categories, which we call "warfarin-responsive index." Warfarin 189-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 16432637-7 2006 Thus, analysis of the combination of VKORC1 and CYP2C9 genotypes should identify warfarin-sensitive patients who require a lower dose of drug, allowing personalized warfarin treatment. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 16924387-0 2006 The high prevalence of the poor and ultrarapid metabolite alleles of CYP2D6, CYP2C9, CYP2C19, CYP3A4, and CYP3A5 in Taiwanese population. ultrarapid 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 16432637-7 2006 Thus, analysis of the combination of VKORC1 and CYP2C9 genotypes should identify warfarin-sensitive patients who require a lower dose of drug, allowing personalized warfarin treatment. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 16719380-0 2006 Determination of CYP2C9-catalyzed diclofenac 4"-hydroxylation by high-performance liquid chromatography. diclofenac 4" 34-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 16719380-1 2006 A reverse-phase, high-performance liquid chromatography method is described for quantification of diclofenac 4"-hydroxylation catalyzed by human liver microsomes or cDNA-expressed CYP2C9. Diclofenac 98-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 16719380-3 2006 This method is applicable to enzymatic studies for determination of CYP2C9-catalyzed diclofenac 4"-hydroxylation activity. Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 16543980-0 2006 A novel sequence variant in exon 7 of CYP2C9 gene (CYP2C9*24) in a patient on warfarin therapy. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 16541193-0 2006 CYP2C9 Ile359Leu polymorphism, plasma irbesartan concentration and acute blood pressure reductions in response to irbesartan treatment in Chinese hypertensive patients. ile359leu 7-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16541193-0 2006 CYP2C9 Ile359Leu polymorphism, plasma irbesartan concentration and acute blood pressure reductions in response to irbesartan treatment in Chinese hypertensive patients. Irbesartan 114-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16541193-1 2006 Our previous study demonstrated that the CYP2C9*3 gene variant was significantly associated with elevated plasma irbesartan concentration and blood pressure decline, assessed by a 4-week follow-up and revisit following daily administration of irbesartan. Irbesartan 113-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 16541193-1 2006 Our previous study demonstrated that the CYP2C9*3 gene variant was significantly associated with elevated plasma irbesartan concentration and blood pressure decline, assessed by a 4-week follow-up and revisit following daily administration of irbesartan. Irbesartan 243-253 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 16541193-8 2006 Subjects with the CYP2C9*1/CYP2C9*3 genotype had significantly higher plasma irbesartan concentrations when compared with those with the CYP2C9*1/CYP2C9*1 genotype (beta +/- SE = 81 +/- 36) and greater DBP response (beta +/- SE = 5.6 +/- 2.5 mmHg) at the 6-h time point after adjusting for important confounders. Irbesartan 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 16541193-8 2006 Subjects with the CYP2C9*1/CYP2C9*3 genotype had significantly higher plasma irbesartan concentrations when compared with those with the CYP2C9*1/CYP2C9*1 genotype (beta +/- SE = 81 +/- 36) and greater DBP response (beta +/- SE = 5.6 +/- 2.5 mmHg) at the 6-h time point after adjusting for important confounders. Irbesartan 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16541193-8 2006 Subjects with the CYP2C9*1/CYP2C9*3 genotype had significantly higher plasma irbesartan concentrations when compared with those with the CYP2C9*1/CYP2C9*1 genotype (beta +/- SE = 81 +/- 36) and greater DBP response (beta +/- SE = 5.6 +/- 2.5 mmHg) at the 6-h time point after adjusting for important confounders. Irbesartan 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16541193-8 2006 Subjects with the CYP2C9*1/CYP2C9*3 genotype had significantly higher plasma irbesartan concentrations when compared with those with the CYP2C9*1/CYP2C9*1 genotype (beta +/- SE = 81 +/- 36) and greater DBP response (beta +/- SE = 5.6 +/- 2.5 mmHg) at the 6-h time point after adjusting for important confounders. Irbesartan 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16541193-9 2006 Our finding suggests that the CYP2C9*3 gene variant significantly alters the plasma concentration and acute DBP response at the 6-h point following irbesartan treatment in Chinese hypertensive patients. Irbesartan 148-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 16543980-0 2006 A novel sequence variant in exon 7 of CYP2C9 gene (CYP2C9*24) in a patient on warfarin therapy. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 16601787-1 2005 Enzyme activities of the CYP enzymes (CYP3A4, CYP2C9 and CYP2A6) were determined using classical substrates (testosterone, diclofenac and coumarin, respectively) as well as with luminogenic or fluorogenic substrates in micromethod arrangement. Testosterone 109-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16188404-0 2005 Down-regulation of astroglial CYP2C, glucocorticoid receptor and constitutive androstane receptor genes in response to cocaine in human U373 MG astrocytoma cells. Cocaine 119-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-35 16188404-2 2005 Cytochromes P450 (CYP), especially the astrocytic members of the CYP2C subfamily may play an important role in the modulation of cerebrovascular functions, by generating vasodilatator metabolites from arachidonic acid (AA). Arachidonic Acid 201-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-70 16188404-3 2005 Our study examined the regulation of CYP2C genes in response to cocaine or amphetamine in the human astrocyte-like U373 MG cells, using reverse transcription-polymerase chain reaction (RT-PCR) and western-blot analysis. Cocaine 64-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-42 16188404-3 2005 Our study examined the regulation of CYP2C genes in response to cocaine or amphetamine in the human astrocyte-like U373 MG cells, using reverse transcription-polymerase chain reaction (RT-PCR) and western-blot analysis. Amphetamine 75-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-42 16188404-4 2005 A treatment for 48h with increasing concentrations of cocaine caused a significant down-regulation of CYP2C8 and CYP2C9 genes and decreased the protein level. Cocaine 54-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 16188404-7 2005 Effects of cocaine on CYP2C were accompanied by a decrease in the GR and CAR gene expression suggesting that these nuclear receptors could be involved in the CYP2C repression by cocaine in the U373 MG cell line. Cocaine 11-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-27 16188404-7 2005 Effects of cocaine on CYP2C were accompanied by a decrease in the GR and CAR gene expression suggesting that these nuclear receptors could be involved in the CYP2C repression by cocaine in the U373 MG cell line. Cocaine 11-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-163 16188404-7 2005 Effects of cocaine on CYP2C were accompanied by a decrease in the GR and CAR gene expression suggesting that these nuclear receptors could be involved in the CYP2C repression by cocaine in the U373 MG cell line. Cocaine 178-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-27 16385662-0 2005 A case of intolerance to warfarin dosing in an intermediate metabolizer of CYP2C9. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 16385662-1 2005 We report a case of intolerance to warfarin dosing due to impaired drug metabolism in a patient heterozygous for the CYP2C9*3 allele. Warfarin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 16188404-7 2005 Effects of cocaine on CYP2C were accompanied by a decrease in the GR and CAR gene expression suggesting that these nuclear receptors could be involved in the CYP2C repression by cocaine in the U373 MG cell line. Cocaine 178-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-163 16601787-1 2005 Enzyme activities of the CYP enzymes (CYP3A4, CYP2C9 and CYP2A6) were determined using classical substrates (testosterone, diclofenac and coumarin, respectively) as well as with luminogenic or fluorogenic substrates in micromethod arrangement. Diclofenac 123-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16305586-1 2005 BACKGROUND: As acetylsalicylic acid is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6) and cytochrome P450 2C9 (CYP2C9), interindividual differences in activity of these enzymes may modulate the effects and side-effects of acetylsalicylic acid. Aspirin 15-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-118 16601787-1 2005 Enzyme activities of the CYP enzymes (CYP3A4, CYP2C9 and CYP2A6) were determined using classical substrates (testosterone, diclofenac and coumarin, respectively) as well as with luminogenic or fluorogenic substrates in micromethod arrangement. coumarin 138-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 16305586-1 2005 BACKGROUND: As acetylsalicylic acid is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6) and cytochrome P450 2C9 (CYP2C9), interindividual differences in activity of these enzymes may modulate the effects and side-effects of acetylsalicylic acid. Aspirin 15-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 16305586-1 2005 BACKGROUND: As acetylsalicylic acid is metabolized by UDP-glucuronosyltransferase 1A6 (UGT1A6) and cytochrome P450 2C9 (CYP2C9), interindividual differences in activity of these enzymes may modulate the effects and side-effects of acetylsalicylic acid. Aspirin 231-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 16305586-2 2005 The objective of this study was to assess whether polymorphisms in UGT1A6 and CYP2C9 genes are related to the prevalence of upper gastrointestinal symptoms in cardiovascular patients using acetylsalicylic acid for secondary prevention of ischaemic heart disease. Aspirin 189-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 16601787-2 2005 The luciferin-based luminogenic substrates for CYP3A4 and CYP2C9 as well as coumarin in micromethod for assay of CYP2A6 activity gave results well comparable with the classical methods with determination of reaction products by the HPLC. D-luciferin 4-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 16111713-1 2005 CYP2C9 is a major P450 2C enzyme, which hydroxylates about 16% of drugs that are in current clinical use and contributes to the metabolism of a number of clinically important substrate drugs such as warfarin. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16183781-1 2005 The present study was conducted to assess a possible in vivo effect of propiverine, an anticholinergic drug to treat urinary incontinence and related disorders, on the activity of intestinal CYP3A4 and of hepatic CYP3A4, CYP2C9, CYP2C19, and CYP1A2. propiverine 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 16192747-3 2005 The risk of such interactions is increased if voriconazole and, much less pronounced caspofungin, are co-administered with drugs which have an inducing or inhibiting effect on the CYP 450 system, primarily on the isoenzymes CYP2C19, CYP2C9 and CYP3A4. Voriconazole 46-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 16338275-2 2005 We aimed to determine the influence of CYP2B6, CYP2C9, and CYP2C19 genetic polymorphism on methadone pharmacokinetics and on the response to treatment. Methadone 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). Oligonucleotides 140-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 16291720-4 2005 In Transwell assays, the migration of endothelial cells pre-exposed to hypoxia to increase CYP expression was abolished by CYP 2C antisense oligonucleotides as well as by the CYP inhibitor MS-PPOH and the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE). 14,15-episulfide eicosatrienoic acid 220-253 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 16291720-7 2005 Matrix metalloprotease (MMP) activity was increased in CYP-2C9-overexpressing cells and correlated with increased invasion through Matrigel-coated Transwell chambers: an effect sensitive to the CYP 2C9 inhibitor sulfaphenazole as well as to EEZE and the MMP inhibitor GM6001. Sulfaphenazole 212-226 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-62 16291720-7 2005 Matrix metalloprotease (MMP) activity was increased in CYP-2C9-overexpressing cells and correlated with increased invasion through Matrigel-coated Transwell chambers: an effect sensitive to the CYP 2C9 inhibitor sulfaphenazole as well as to EEZE and the MMP inhibitor GM6001. N-(2(R)-2-(hydroxamidocarbonylmethyl)-4-methylpentanoyl)-L-tryptophan methylamide 268-274 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-62 16237546-9 2005 The minor role of CYP2C9 in 4"-hydroxy-PPC formation and the effect of CYP2C9 genotype for (S)-6- and (S)-7-hydroxy-PPC were confirmed. S 6 91-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16237546-9 2005 The minor role of CYP2C9 in 4"-hydroxy-PPC formation and the effect of CYP2C9 genotype for (S)-6- and (S)-7-hydroxy-PPC were confirmed. (s)-7-hydroxy-ppc 102-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. Fluoxetine 57-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 16236141-3 2005 In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. norfluoxetine 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 16236141-9 2005 Among homozygous extensive metabolizers for CYP2D6, patients homozygous for CYP2C9*1 had lower dose-normalized R-fluoxetine concentrations and lower active moiety levels compared with those carrying detrimental CYP2C9 alleles (P<0.05). (R)-Fluoxetine 111-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 16081671-2 2005 There was an excellent correlation between IC(50, apparent) values determined using diclofenac and naproxen as CYP2C9 substrates (r2 = 0.82, p < 0.0001), with values being generally higher in the naproxen assay. Naproxen 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 16236141-10 2005 These results suggest that CYP2D6 and CYP2C9 polymorphisms contribute to the interindividual variability in fluoxetine pharmacokinetics at steady-state. Fluoxetine 108-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 16321620-1 2005 OBJECTIVE: Our objective was to prospectively study the impact of CYP2C9 polymorphism (*2 and *3) on the risk of overanticoagulation during the induction phase of warfarin therapy. Warfarin 163-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 16321620-8 2005 CONCLUSIONS: The CYP2C9*2 and *3 single-nucleotide polymorphisms significantly increase the risk of overanticoagulation during the first 2 weeks of warfarin treatment, with increased INR levels evident after only 4 days" treatment in *2 carriers. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 16321620-9 2005 Our prospective data are consistent with results from previous retrospective studies and indicate that CYP2C9 genotyping may be a means of improving safety during warfarin induction. Warfarin 163-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 16236040-1 2005 AIMS: Dexloxiglumide is a new CCK(1) receptor antagonist under investigation for treatment of functional gastrointestinal disorders and is metabolized by CYP3A4 and CYP2C9. dexloxiglumide 6-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 16303885-0 2005 Impact of age, CYP2C9 genotype and concomitant medication on the rate of rise for prothrombin time during the first 30 days of warfarin therapy. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16118328-6 2005 Therefore, a number of COX inhibitors were surveyed and it was determined that CYP2C9 plays a relatively minor role in the overall clearance (<or=20% of the dose) of sulindac, naproxen, ketoprofen, diclofenac, rofecoxib, and etoricoxib. Sulindac 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 16118328-6 2005 Therefore, a number of COX inhibitors were surveyed and it was determined that CYP2C9 plays a relatively minor role in the overall clearance (<or=20% of the dose) of sulindac, naproxen, ketoprofen, diclofenac, rofecoxib, and etoricoxib. Diclofenac 201-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Ibuprofen 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Indomethacin 79-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Flurbiprofen 93-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Celecoxib 107-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. valdecoxib 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. lornoxicam 130-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Meloxicam 153-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16118328-8 2005 In contrast, CYP2C9 genotype is expected to impact the clearance of ibuprofen, indomethacin, flurbiprofen, celecoxib, valdecoxib, lornoxicam, tenoxicam, meloxicam, and piroxicam. Piroxicam 168-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16248835-5 2005 Further study conducted in cDNA-expressed cytochrome P450s (CYPs) demonstrates that the inhibition of the in-vitro biotransformation of warfarin enantiomers by omeprazole is attributed to its inhibitory effect on the activities of CYP1A2, CYP3A4, CYP2C9 and CYP2C19. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 247-253 16112652-0 2005 CYP2C-catalyzed delta9-tetrahydrocannabinol metabolism: kinetics, pharmacogenetics and interaction with phenytoin. Dronabinol 16-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-5 16112652-0 2005 CYP2C-catalyzed delta9-tetrahydrocannabinol metabolism: kinetics, pharmacogenetics and interaction with phenytoin. Phenytoin 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-5 16220110-2 2005 Among them, phenytoin has been used as a probe to determine CYP2C9 phenotype by measuring the urinary excretion of its major metabolite, S-enantiomer of 5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH). Phenytoin 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 16220110-2 2005 Among them, phenytoin has been used as a probe to determine CYP2C9 phenotype by measuring the urinary excretion of its major metabolite, S-enantiomer of 5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH). hydroxyphenytoin 153-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 16220110-2 2005 Among them, phenytoin has been used as a probe to determine CYP2C9 phenotype by measuring the urinary excretion of its major metabolite, S-enantiomer of 5-(4-hydroxyphenyl)-5-phenylhydantoin (p-HPPH). p-hpph 192-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 16220110-10 2005 On the other hand, only CYP2C9 was predictive for the urinary excretion of (S)-p-HPPH and PMR (r=0.21, P=0.001 and r=0.25, P<0.001, respectively). (s)-p-hpph 75-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 16220110-11 2005 Furthermore, significant relation was found between urinary excretion of (R)-p-HPPH and CYP2C9 genotype (P=0.035) and levels significantly increased in the genotype order: CYP2C9*1/*9, CYP2C9*1/*1, CYP2C9*9/*11, CYP2C9*1/*8 and CYP2C9*1/*5 (P<0.001, Jonckheere-Terpstra test). (r)-p-hpph 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 16220110-12 2005 In summary, the present study demonstrates that, in a Black population, CYP2C9*5, *6, *8 and *11 variants, but not CYP2C9*9, are associated with a decreased phenytoin metabolism. Phenytoin 157-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 15947090-5 2005 The mean warfarin daily dose requirement was highest in CYP2C9 homozygous wild-type patients, compared with those with the variant *2 and *3 alleles (P < .001) and highest in patients with the VKORC1 (position -1639) GG genotype compared with those with the GA genotype and the AA genotype (P < .001). Warfarin 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 15947090-7 2005 Age, height, and CYP2C9 genotype significantly contributed to S-warfarin and total warfarin clearance, whereas only age and body size significantly contributed to R-warfarin clearance. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 15947090-7 2005 Age, height, and CYP2C9 genotype significantly contributed to S-warfarin and total warfarin clearance, whereas only age and body size significantly contributed to R-warfarin clearance. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 15947090-7 2005 Age, height, and CYP2C9 genotype significantly contributed to S-warfarin and total warfarin clearance, whereas only age and body size significantly contributed to R-warfarin clearance. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 15947090-8 2005 The multivariate regression model including the variables of age, CYP2C9 and VKORC1 genotype, and height produced the best model for estimating warfarin dose (R2 = 55%). Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 16198655-0 2005 Influence of CYP2C9 genotypes on the pharmacokinetics and pharmacodynamics of piroxicam. Piroxicam 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16198655-1 2005 OBJECTIVE: Our objective was to evaluate the influence of cytochrome P450 (CYP) 2C9 polymorphisms on the pharmacokinetics and pharmacodynamics of the nonsteroidal anti-inflammatory drug piroxicam. Piroxicam 186-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-83 16198655-8 2005 CONCLUSION: Piroxicam"s oral clearance was impaired and its inhibitory effect on cyclooxygenase 1 activity was increased in CYP2C9*1/*2 or CYP2C9*1/*3 individuals, as compared with CYP2C 9*1 homozygous individuals. Piroxicam 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 16198655-8 2005 CONCLUSION: Piroxicam"s oral clearance was impaired and its inhibitory effect on cyclooxygenase 1 activity was increased in CYP2C9*1/*2 or CYP2C9*1/*3 individuals, as compared with CYP2C 9*1 homozygous individuals. Piroxicam 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 16198655-8 2005 CONCLUSION: Piroxicam"s oral clearance was impaired and its inhibitory effect on cyclooxygenase 1 activity was increased in CYP2C9*1/*2 or CYP2C9*1/*3 individuals, as compared with CYP2C 9*1 homozygous individuals. Piroxicam 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-188 16198656-0 2005 CYP2C9, but not CYP2C19, polymorphisms affect the pharmacokinetics and pharmacodynamics of glyburide in Chinese subjects. Glyburide 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16198656-1 2005 BACKGROUND: Although cytochrome P450 (CYP) 2C9 was thought to be the main pathway for glyburide (INN, glibenclamide) metabolism in vivo, studies in vitro indicated that CYP2C19 had a more dominant effect. Glyburide 86-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-46 16198656-1 2005 BACKGROUND: Although cytochrome P450 (CYP) 2C9 was thought to be the main pathway for glyburide (INN, glibenclamide) metabolism in vivo, studies in vitro indicated that CYP2C19 had a more dominant effect. Glyburide 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-46 16198656-11 2005 CONCLUSION: CYP2C9, but not CYP2C19, polymorphism appears to exert a dominant influence on glyburide pharmacokinetics and pharmacodynamics in vivo. Glyburide 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16248835-5 2005 Further study conducted in cDNA-expressed cytochrome P450s (CYPs) demonstrates that the inhibition of the in-vitro biotransformation of warfarin enantiomers by omeprazole is attributed to its inhibitory effect on the activities of CYP1A2, CYP3A4, CYP2C9 and CYP2C19. Omeprazole 160-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 247-253 16112652-4 2005 The present study was designed to address the kinetics and pharmacogenetics of CYP2C-mediated metabolism of (delta9)-THC by studying its metabolism in human liver microsomes and expressed enzymes. Dronabinol 108-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-84 16112652-5 2005 Expressed CYP2C9.1 exhibited high affinity for the hydroxylation of delta9-THC (apparent Km, 2 microM), similar to that observed in human liver microsomes (apparent Km 0.8 microM). Dronabinol 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 16112652-7 2005 Given the high affinity of CYP2C9 for the hydroxylation of delta9-THC, we evaluated the potential for an interaction between delta9-THC, 11-hydroxy-delta9-THC, or 11-nor-9-carboxy-delta9-THC and the CYP2C9 substrate, phenytoin. Dronabinol 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16112652-7 2005 Given the high affinity of CYP2C9 for the hydroxylation of delta9-THC, we evaluated the potential for an interaction between delta9-THC, 11-hydroxy-delta9-THC, or 11-nor-9-carboxy-delta9-THC and the CYP2C9 substrate, phenytoin. 11-nor-delta(9)-tetrahydrocannabinol-9-carboxylic acid 163-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 199-205 16112652-8 2005 Surprisingly, delta9-THC increased the rate of phenytoin hydroxylation in human liver microsomes and expressed CYP2C9 enzyme. Dronabinol 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 16094537-0 2005 CYP2C9*3 allelic variant is associated with metabolism of irbesartan in Chinese population. Irbesartan 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16094537-2 2005 The objective of this study was to test the presence of the CYP2C9*3 allelic variant in the Chinese population and to investigate the association of this variant with both metabolism and therapeutic efficacy of irbesartan on essential hypertension. Irbesartan 211-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 16094537-9 2005 Multiple linear regression analyses revealed that the CYP2C9*3 allele carriers had significantly higher irbesartan concentrations in plasma at 6 h (Taihu: P < 0.0001; Dongzhi: P = 0.03) and 24 h (Taihu: P < 0.0001; Dongzhi: P = 0.00013) after dosing. Irbesartan 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 16094537-11 2005 CONCLUSION: Our study suggests that the CYP2C9*3 plays an important role in the metabolism of irbesartan and/or is in linkage disequilibrium with another potential CYP2C9 allele, both of which possibly modify the pharmacokinetics of irbesartan. Irbesartan 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 16094537-11 2005 CONCLUSION: Our study suggests that the CYP2C9*3 plays an important role in the metabolism of irbesartan and/or is in linkage disequilibrium with another potential CYP2C9 allele, both of which possibly modify the pharmacokinetics of irbesartan. Irbesartan 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 16094537-11 2005 CONCLUSION: Our study suggests that the CYP2C9*3 plays an important role in the metabolism of irbesartan and/or is in linkage disequilibrium with another potential CYP2C9 allele, both of which possibly modify the pharmacokinetics of irbesartan. Irbesartan 233-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 16094537-11 2005 CONCLUSION: Our study suggests that the CYP2C9*3 plays an important role in the metabolism of irbesartan and/or is in linkage disequilibrium with another potential CYP2C9 allele, both of which possibly modify the pharmacokinetics of irbesartan. Irbesartan 233-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 16141794-9 2005 In multivariate analysis, VKORC1 and CYP2C9 explained 31% and 7.9% of the variability in warfarin dose, respectively. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 16205037-3 2005 Fluconazole, micafungin, and voriconazole have lower inhibitory effects on CYP3A4 activities than itraconazole and miconazole, and IC(50) and/or K(i) values against CYP2C9 and CYP2C19 activities are the lowest for miconazole, followed by voriconazole and fluconazole. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 16270629-7 2005 Since hereditary pharmacodynamic (VKORC1) and pharmacokinetic (CYP2C9) factors account for up to 50% of the inter-individual variability of the warfarin response, these genetic markers may serve as clinically relevant predictors of warfarin dosing in future studies. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 16270629-7 2005 Since hereditary pharmacodynamic (VKORC1) and pharmacokinetic (CYP2C9) factors account for up to 50% of the inter-individual variability of the warfarin response, these genetic markers may serve as clinically relevant predictors of warfarin dosing in future studies. Warfarin 232-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 16205037-6 2005 Miconazole is a potent inhibitor of all CYPs investigated in vitro, although there are few detailed studies on the clinical significance of this except for CYP2C9. Miconazole 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 16205037-3 2005 Fluconazole, micafungin, and voriconazole have lower inhibitory effects on CYP3A4 activities than itraconazole and miconazole, and IC(50) and/or K(i) values against CYP2C9 and CYP2C19 activities are the lowest for miconazole, followed by voriconazole and fluconazole. Micafungin 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 16205037-3 2005 Fluconazole, micafungin, and voriconazole have lower inhibitory effects on CYP3A4 activities than itraconazole and miconazole, and IC(50) and/or K(i) values against CYP2C9 and CYP2C19 activities are the lowest for miconazole, followed by voriconazole and fluconazole. Voriconazole 29-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 16205037-4 2005 In in vivo pharmacokinetic studies, it is well known that itraconazole is a potent clinically important inhibitor of the clearance of CYP3A4 substrates, and fluconazole and voriconazole are reported to increase the blood or plasma concentrations of not only midazolam and cyclosporine (CYP3A4 substrates) but also of phenytoin (CYP2C9 substrate) and/or omeprazole (CYP2C19/CYP3A4 substrate). Itraconazole 58-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 328-334 16205037-4 2005 In in vivo pharmacokinetic studies, it is well known that itraconazole is a potent clinically important inhibitor of the clearance of CYP3A4 substrates, and fluconazole and voriconazole are reported to increase the blood or plasma concentrations of not only midazolam and cyclosporine (CYP3A4 substrates) but also of phenytoin (CYP2C9 substrate) and/or omeprazole (CYP2C19/CYP3A4 substrate). Fluconazole 157-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 328-334 16408808-4 2005 RESULTS: Imrecoxib is metabolized by CYP2C9, CYP2D6 and CYP3A4, with the rate of 62.5%, 21.1% and 16.4%, respectively. Imrecoxib 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 16205037-4 2005 In in vivo pharmacokinetic studies, it is well known that itraconazole is a potent clinically important inhibitor of the clearance of CYP3A4 substrates, and fluconazole and voriconazole are reported to increase the blood or plasma concentrations of not only midazolam and cyclosporine (CYP3A4 substrates) but also of phenytoin (CYP2C9 substrate) and/or omeprazole (CYP2C19/CYP3A4 substrate). Voriconazole 173-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 328-334 16141567-1 2005 The effects of five antifungal drugs, fluconazole, itraconazole, micafungin, miconazole, and voriconazole, on cytochrome P450 (CYP) 2C9-mediated tolbutamide hydroxylation, CYP2C19-mediated S-mephenytoin 4"-hydroxylation, and CYP3A4-mediated nifedipine oxidation activities in human liver microsomes were compared. Miconazole 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-135 16408808-5 2005 CONCLUSION: CYP2C9 is the major enzyme involved in imrecoxib hydroxylation metabolism. Imrecoxib 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16014372-7 2005 The mechanism of action for the interaction is not clear, but may be related to down-regulation of CYP2C9 by capecitabine or its metabolites or a pharmacodynamic interaction with warfarin. Capecitabine 109-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 16197363-8 2005 Recent drug-drug interaction studies have demonstrated that meloxicam interacts with some medications, including cholestyramine, lithium and some inhibitors of cytochrome P450 -2C9 and -3A4. Meloxicam 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-180 16141567-1 2005 The effects of five antifungal drugs, fluconazole, itraconazole, micafungin, miconazole, and voriconazole, on cytochrome P450 (CYP) 2C9-mediated tolbutamide hydroxylation, CYP2C19-mediated S-mephenytoin 4"-hydroxylation, and CYP3A4-mediated nifedipine oxidation activities in human liver microsomes were compared. Voriconazole 93-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-135 16141567-1 2005 The effects of five antifungal drugs, fluconazole, itraconazole, micafungin, miconazole, and voriconazole, on cytochrome P450 (CYP) 2C9-mediated tolbutamide hydroxylation, CYP2C19-mediated S-mephenytoin 4"-hydroxylation, and CYP3A4-mediated nifedipine oxidation activities in human liver microsomes were compared. Tolbutamide 145-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-135 15924351-1 2005 Microsomal epoxide hydrolase (mEH) and cytochrome P450 2C9 (CYP2C9) are involved in the bioactivation and detoxification of polycyclic aromatic hydrocarbons (PAHs) derived from tobacco smoke. Polycyclic Aromatic Hydrocarbons 124-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 15856231-8 2005 The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. Paclitaxel 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-88 15856231-8 2005 The effect of paclitaxel treatment on hepatic expression of PGP and P450 isoforms (CYP2C and CYP3A) was determined to elucidate the mechanism by which paclitaxel disposition is altered by previous drug exposure. Paclitaxel 151-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-88 15856231-11 2005 Activity and protein levels for CYP2C in liver were elevated at 24 and 96 h after paclitaxel dosing. Paclitaxel 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-37 15856231-12 2005 Cremophor EL, a carrier solvent for paclitaxel, also caused elevated CYP2C activity. cremophor EL 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-74 15856231-12 2005 Cremophor EL, a carrier solvent for paclitaxel, also caused elevated CYP2C activity. Paclitaxel 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-74 15919766-2 2005 Recent studies in our laboratory have shown that the nuclear receptor pregnane X receptor (PXR) is important in the transcriptional activation of the CYP2C9 promoter by drugs such as rifampicin and that the essential element is a constitutive androstane receptor (CAR)/PXR site -1839 bp upstream of the translation start site. Rifampin 183-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 15919766-8 2005 Mutation of the two HNF4alpha binding sites differentially prevented up-regulation of CYP2C9 promoter by both CAR as well as HNF4alpha, synergy between the two receptors, and essentially abolished induction by rifampicin in HepG2 cells transfected with PXR. Rifampin 210-220 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 16153401-0 2005 Cytochrome P450 2C9 genotype: impact on celecoxib safety and pharmacokinetics in a pediatric patient. Celecoxib 40-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15924351-1 2005 Microsomal epoxide hydrolase (mEH) and cytochrome P450 2C9 (CYP2C9) are involved in the bioactivation and detoxification of polycyclic aromatic hydrocarbons (PAHs) derived from tobacco smoke. Polycyclic Aromatic Hydrocarbons 158-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 15955872-0 2005 CYP2C9 genotype-dependent effects on in vitro drug-drug interactions: switching of benzbromarone effect from inhibition to activation in the CYP2C9.3 variant. Benzbromarone 83-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16308280-2 2005 CYP2C9*13 is an allele identified in a Chinese poor metabolizer of lornoxicam which has a Leu90Pro amino acid substitution. lornoxicam 67-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16308280-3 2005 This paper reports on a study aimed at comparing the catalytic properties of CYP2C9*13 with those of the wild-type CYP2C9*1 and mutant CYP2C9*3 (Ile359Leu) in the COS-7 expression system using various substrates. carbonyl sulfide 163-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 16308280-3 2005 This paper reports on a study aimed at comparing the catalytic properties of CYP2C9*13 with those of the wild-type CYP2C9*1 and mutant CYP2C9*3 (Ile359Leu) in the COS-7 expression system using various substrates. carbonyl sulfide 163-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 16308280-3 2005 This paper reports on a study aimed at comparing the catalytic properties of CYP2C9*13 with those of the wild-type CYP2C9*1 and mutant CYP2C9*3 (Ile359Leu) in the COS-7 expression system using various substrates. carbonyl sulfide 163-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 16308280-4 2005 CYP2C9*3 and *13 produced far lower luminescence than CYP2C9*1 in luciferin H metabolism. luciferin h 66-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16308280-4 2005 CYP2C9*3 and *13 produced far lower luminescence than CYP2C9*1 in luciferin H metabolism. luciferin h 66-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 16308280-5 2005 CYP2C9*13 exhibited an 11-fold increase in Km but no change in Vmax with tolbutamide as the substrate, a five-fold increase in Km and an 88.8% reduction in Vmax with diclofenac. Diclofenac 166-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. Nateglinide 161-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 16042675-11 2005 The findings suggest that in vivo gemfibrozil, probably due to its metabolites, is a much more potent inhibitor of CYP2C8 than of CYP2C9. Gemfibrozil 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 16268502-0 2005 [Influence of cytochrom P450 CYP2C9 polymorphism on the pharmacokinetics of tolbutamide metabolism using oligonucleotide genotyping microarray]. Tolbutamide 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 16268502-0 2005 [Influence of cytochrom P450 CYP2C9 polymorphism on the pharmacokinetics of tolbutamide metabolism using oligonucleotide genotyping microarray]. Oligonucleotides 105-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 16268502-1 2005 AIM: To investigate the influence of cytochrom P450 CYP2C9 polymorphism on the pharmacokinetics of tolbutamide. Tolbutamide 99-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 16268502-9 2005 Pharmacokinetic outcome showed that the individuals with CYP2C9 *1/*3 or CYP2C9 *3/*3 had slower metabolic elimination of tolbutamide than those with CYP2C9 *1/*1. Tolbutamide 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16268502-9 2005 Pharmacokinetic outcome showed that the individuals with CYP2C9 *1/*3 or CYP2C9 *3/*3 had slower metabolic elimination of tolbutamide than those with CYP2C9 *1/*1. Tolbutamide 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 16268502-9 2005 Pharmacokinetic outcome showed that the individuals with CYP2C9 *1/*3 or CYP2C9 *3/*3 had slower metabolic elimination of tolbutamide than those with CYP2C9 *1/*1. Tolbutamide 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 16268502-10 2005 CONCLUSION: CYP2C9 genetic polymorphism has a significant influence on the pharmacokinetics of tolbutamide. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16278191-3 2005 The formation of norketobemidone from ketobemidone (1 microM) correlated best with CYP2C9 activity, measured as losartan oxidation (rs = 0.82, n = 19, p < 0.001), but there was also a strong correlation with CYP3A4 activity. norketobemidone 17-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 16278191-3 2005 The formation of norketobemidone from ketobemidone (1 microM) correlated best with CYP2C9 activity, measured as losartan oxidation (rs = 0.82, n = 19, p < 0.001), but there was also a strong correlation with CYP3A4 activity. ketobemidone 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 16278191-3 2005 The formation of norketobemidone from ketobemidone (1 microM) correlated best with CYP2C9 activity, measured as losartan oxidation (rs = 0.82, n = 19, p < 0.001), but there was also a strong correlation with CYP3A4 activity. Losartan 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 16278191-5 2005 Inhibition studies confirmed the involvement of CYP2C9 and CTP3A4 in the formation of norketobemidone. norketobemidone 86-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 16278191-6 2005 The formation rate of norketobemidone was three times higher in the CYP2C9*1*1 genotype group compared with the CYP2C9*1*2, CYP2C9*1*3 and CYP2C9*3*3 genotypes (p < 0.01). norketobemidone 22-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 16278191-6 2005 The formation rate of norketobemidone was three times higher in the CYP2C9*1*1 genotype group compared with the CYP2C9*1*2, CYP2C9*1*3 and CYP2C9*3*3 genotypes (p < 0.01). norketobemidone 22-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 16278191-6 2005 The formation rate of norketobemidone was three times higher in the CYP2C9*1*1 genotype group compared with the CYP2C9*1*2, CYP2C9*1*3 and CYP2C9*3*3 genotypes (p < 0.01). norketobemidone 22-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 16278191-6 2005 The formation rate of norketobemidone was three times higher in the CYP2C9*1*1 genotype group compared with the CYP2C9*1*2, CYP2C9*1*3 and CYP2C9*3*3 genotypes (p < 0.01). norketobemidone 22-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 16278191-8 2005 The data suggest that CYP2C9 and CYP3A4 play a major role in the formation of norketobemidone at clinically relevant concentrations. norketobemidone 78-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 15790782-0 2005 Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) genotypes as determinants of acenocoumarol sensitivity. Acenocoumarol 99-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15790782-0 2005 Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) genotypes as determinants of acenocoumarol sensitivity. Acenocoumarol 99-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15955872-3 2005 Although benzbromarone inhibited CYP2C9.1 activity as expected, CYP2C9.3-mediated flurbiprofen 4"-hydroxylation was activated in the presence of benzbromarone. Flurbiprofen 82-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 15955872-3 2005 Although benzbromarone inhibited CYP2C9.1 activity as expected, CYP2C9.3-mediated flurbiprofen 4"-hydroxylation was activated in the presence of benzbromarone. Benzbromarone 145-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 16342679-0 2005 [Impact of cytochrome P450 CYP2C9 variant allele CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam in Chinese subjects]. Glyburide 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16342679-0 2005 [Impact of cytochrome P450 CYP2C9 variant allele CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam in Chinese subjects]. Glyburide 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 16342679-0 2005 [Impact of cytochrome P450 CYP2C9 variant allele CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam in Chinese subjects]. lornoxicam 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 16342679-0 2005 [Impact of cytochrome P450 CYP2C9 variant allele CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam in Chinese subjects]. lornoxicam 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 16342679-1 2005 AIM: To investigate the impact of CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam. Glyburide 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 16342679-1 2005 AIM: To investigate the impact of CYP2C9 * 3 on the pharmacokinetics of glibenclamide and lornoxicam. lornoxicam 90-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 16342679-5 2005 RESULTS: After a single oral dose of 2.5 mg glibenclamide, C(max) was (70.0 +/- 11.5) microg x L(-1) in CYP2C9 * 1/ * 3 subjects and (51.9 +/- 12.3) microg x L(-1) in * 1/ *1 subjects. Glyburide 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 16342679-7 2005 Statistic analysis results indicated that glibenclamide AUC(0-infinity) was significantly higher (1.5-fold) and subsequently CL/F was significantly lower (40%) in CYP2C9 * 1/ * 3 subjects than those in * 1/ * 1 subjects (P < 0.01). Glyburide 42-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 16342679-7 2005 Statistic analysis results indicated that glibenclamide AUC(0-infinity) was significantly higher (1.5-fold) and subsequently CL/F was significantly lower (40%) in CYP2C9 * 1/ * 3 subjects than those in * 1/ * 1 subjects (P < 0.01). Fluorine 128-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 16342679-8 2005 After a single oral dose of 8 mg lornoxicam, C(max) was (1.54 +/- 0.24) mg x L(-1) in CYP2C9 * 1/ * 3 subjects and (1.19 +/- 0.37) mg x L(-1) in * 1/ * 1 subjects. lornoxicam 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 16342679-11 2005 2-fold) and subsequently CL/F was significantly lower (55% ) in CYP2C9 * 1/ * 3 subjects than those in * 1/ * 1 subjects (P < 0.001). Fluorine 28-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 16342679-12 2005 CONCLUSION: CYP2C9 * 3 greatly affects both the pharmacokinetic profiles of glibenclamide and lornoxicam. Glyburide 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16342679-12 2005 CONCLUSION: CYP2C9 * 3 greatly affects both the pharmacokinetic profiles of glibenclamide and lornoxicam. lornoxicam 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 16342679-13 2005 The elimination of these drugs significantly decreased in subjects with CYP2C9 * 1/ * 3 genotype, especially lornoxicam. lornoxicam 109-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 16042675-0 2005 Coadministration of gemfibrozil and itraconazole has only a minor effect on the pharmacokinetics of the CYP2C9 and CYP3A4 substrate nateglinide. Gemfibrozil 20-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 16042675-0 2005 Coadministration of gemfibrozil and itraconazole has only a minor effect on the pharmacokinetics of the CYP2C9 and CYP3A4 substrate nateglinide. Itraconazole 36-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 16042675-2 2005 In vitro, gemfibrozil is a more potent inhibitor of CYP2C9 than of CYP2C8. Gemfibrozil 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. Gemfibrozil 46-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. Itraconazole 58-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 16042675-3 2005 Our aim was to investigate the effects of the gemfibrozil-itraconazole combination on the pharmacokinetics and pharmacodynamics of another meglitinide analogue, nateglinide, which is metabolized by CYP2C9 and CYP3A4. meglitinide 139-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 16160068-0 2005 A prospective, randomized pilot trial of model-based warfarin dose initiation using CYP2C9 genotype and clinical data. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 16160068-1 2005 BACKGROUND: Rapid genetic screening for cytochrome P450 (CYP) 2C9 variants may play a role in improving the efficacy and safety of warfarin in individuals with CYP2C9 variants. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-65 16160068-1 2005 BACKGROUND: Rapid genetic screening for cytochrome P450 (CYP) 2C9 variants may play a role in improving the efficacy and safety of warfarin in individuals with CYP2C9 variants. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 16160068-2 2005 The feasibility of prospective CYP2C9 model-based warfarin dosing has not yet been assessed. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 16160068-3 2005 OBJECTIVES: To evaluate the feasibility of applying a CYP2C9 gene-based warfarin dosing model in clinical practice. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 16160068-20 2005 CONCLUSIONS: Prospective application of a multivariate CYP2C9 gene-based warfarin dosing model is feasible. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 15914520-10 2005 Voriconazole is metabolized primarily by CYP2C19, as well as CYP2C9 and CYP3A4. Voriconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 15963101-0 2005 Association between CYP2C9 slow metabolizer genotypes and severe hypoglycaemia on medication with sulphonylurea hypoglycaemic agents. Sulfonylurea Compounds 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 15963101-1 2005 AIMS: The genetically polymorphic cytochrome P450 (CYP) enzyme CYP2C9 metabolizes most sulphonylurea oral hypoglycaemic agents. Sulfonylurea Compounds 87-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15843486-1 2005 Recently, two new classes of reversible inhibitors, the benzbromarones (BZBRs) and the N-3 substituted phenobarbitals (PBs), were used to study the active site characteristics of CYP2C9 and 2C19, respectively. Benzbromarone 56-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15843486-1 2005 Recently, two new classes of reversible inhibitors, the benzbromarones (BZBRs) and the N-3 substituted phenobarbitals (PBs), were used to study the active site characteristics of CYP2C9 and 2C19, respectively. Benzbromarone 72-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15843486-1 2005 Recently, two new classes of reversible inhibitors, the benzbromarones (BZBRs) and the N-3 substituted phenobarbitals (PBs), were used to study the active site characteristics of CYP2C9 and 2C19, respectively. n-3 substituted phenobarbitals 87-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15843486-1 2005 Recently, two new classes of reversible inhibitors, the benzbromarones (BZBRs) and the N-3 substituted phenobarbitals (PBs), were used to study the active site characteristics of CYP2C9 and 2C19, respectively. Phenobarbital 119-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15843491-6 2005 Significant (p < 0.05) inhibition by selective P450 isoform inhibitor sulfaphenazole (2.1 microM; CYP2C9) indicated a role for CYP2C9 in the formation of the hydroxylamine. Sulfaphenazole 73-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 15843491-6 2005 Significant (p < 0.05) inhibition by selective P450 isoform inhibitor sulfaphenazole (2.1 microM; CYP2C9) indicated a role for CYP2C9 in the formation of the hydroxylamine. Sulfaphenazole 73-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 15843491-6 2005 Significant (p < 0.05) inhibition by selective P450 isoform inhibitor sulfaphenazole (2.1 microM; CYP2C9) indicated a role for CYP2C9 in the formation of the hydroxylamine. Hydroxylamine 161-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 15843491-6 2005 Significant (p < 0.05) inhibition by selective P450 isoform inhibitor sulfaphenazole (2.1 microM; CYP2C9) indicated a role for CYP2C9 in the formation of the hydroxylamine. Hydroxylamine 161-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 15843491-8 2005 Fluconazole (CYP2C9/19 and CYP3A4 inhibitor at clinical concentrations) inhibited hydroxylamine formation, with one-enzyme model K(i) estimates ranging from 9 to 40 microM. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15843491-8 2005 Fluconazole (CYP2C9/19 and CYP3A4 inhibitor at clinical concentrations) inhibited hydroxylamine formation, with one-enzyme model K(i) estimates ranging from 9 to 40 microM. Hydroxylamine 82-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15952022-10 2005 APOE genotype explains 6% of warfarin dose variance among CYP2C9 extensive metabolisers (analysis of variance, P=0.009). Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15983826-14 2005 CONCLUSION: These data are consistent with a significant induction of CYP2C9 metabolism of S(-) warfarin by aprepitant. Sulfur 6-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 15983826-14 2005 CONCLUSION: These data are consistent with a significant induction of CYP2C9 metabolism of S(-) warfarin by aprepitant. Warfarin 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 15888487-4 2005 We identified three CYP2C9 variants, CYP2C9*3, T299A and P382L, in four warfarin-sensitive patients. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 15888487-4 2005 We identified three CYP2C9 variants, CYP2C9*3, T299A and P382L, in four warfarin-sensitive patients. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 15970795-0 2005 In-vitro and in-vivo effects of the CYP2C9*11 polymorphism on warfarin metabolism and dose. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 15970795-1 2005 OBJECTIVE: To determine the in-vitro and in-vivo effects of the CYP2C9*11 polymorphism on (S)-warfarin metabolism. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 15970795-7 2005 Metabolic studies demonstrated that functional CYP2C9.11 possessed similar (S)-warfarin hydroxylation regioselectivity and modestly reduced catalytic efficiency relative to the wild-type enzyme. Warfarin 75-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 15970795-8 2005 CONCLUSIONS: In-vivo reduction in CYP2C9 (S)-warfarin activity due to the CYP2C9*11 polymorphism may largely be a consequence of decreased enzyme stability resulting in compromised expression of holo-enzyme. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 15970795-8 2005 CONCLUSIONS: In-vivo reduction in CYP2C9 (S)-warfarin activity due to the CYP2C9*11 polymorphism may largely be a consequence of decreased enzyme stability resulting in compromised expression of holo-enzyme. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15970795-9 2005 Increased enzyme lability of CYP2C9.11 may be related to improper folding due to the disruption of conserved salt-bridge and hydrogen bonding contacts in the loop region between the J and J" helices of the protein. Hydrogen 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 15955872-0 2005 CYP2C9 genotype-dependent effects on in vitro drug-drug interactions: switching of benzbromarone effect from inhibition to activation in the CYP2C9.3 variant. Benzbromarone 83-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15955872-2 2005 To examine this possibility, the effect of the potent CYP2C9 inhibitor, benzbromarone, was studied with regard to CYP2C9.1- and CYP2C9.3-mediated flurbiprofen metabolism to evaluate whether the variant enzyme exhibits differential inhibition kinetics. Benzbromarone 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 15955872-2 2005 To examine this possibility, the effect of the potent CYP2C9 inhibitor, benzbromarone, was studied with regard to CYP2C9.1- and CYP2C9.3-mediated flurbiprofen metabolism to evaluate whether the variant enzyme exhibits differential inhibition kinetics. Benzbromarone 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 15955872-2 2005 To examine this possibility, the effect of the potent CYP2C9 inhibitor, benzbromarone, was studied with regard to CYP2C9.1- and CYP2C9.3-mediated flurbiprofen metabolism to evaluate whether the variant enzyme exhibits differential inhibition kinetics. Benzbromarone 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 15955872-3 2005 Although benzbromarone inhibited CYP2C9.1 activity as expected, CYP2C9.3-mediated flurbiprofen 4"-hydroxylation was activated in the presence of benzbromarone. Benzbromarone 9-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15764711-0 2005 Role of CYP2C9 and its variants (CYP2C9*3 and CYP2C9*13) in the metabolism of lornoxicam in humans. lornoxicam 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 15764713-9 2005 Using cDNA-expressed cytochrome P450 isoenzymes, it was determined that several isoforms contributed to the metabolism of bortezomib, including CYP3A4, CYP2C19, CYP1A2, CYP2D6, and CYP2C9. Bortezomib 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 15764711-0 2005 Role of CYP2C9 and its variants (CYP2C9*3 and CYP2C9*13) in the metabolism of lornoxicam in humans. lornoxicam 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15764717-0 2005 Detection of a novel reactive metabolite of diclofenac: evidence for CYP2C9-mediated bioactivation via arene oxides. Diclofenac 44-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 15764711-0 2005 Role of CYP2C9 and its variants (CYP2C9*3 and CYP2C9*13) in the metabolism of lornoxicam in humans. lornoxicam 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15764711-2 2005 Recently, we identified a new CYP2C9 allele with a Leu90Pro mutation in a Chinese poor metabolizer of lornoxicam [Si D, Guo Y, Zhang Y, Yang L, Zhou H, and Zhong D (2004) Pharmacogenetics 14:465-469]. lornoxicam 102-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15764717-0 2005 Detection of a novel reactive metabolite of diclofenac: evidence for CYP2C9-mediated bioactivation via arene oxides. arene oxides 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 15764711-4 2005 To examine enzymatic activity of the CYP2C9*13 allele, kinetic parameters for lornoxicam 5"-hydroxylation were determined in COS-7 cells transiently transfected with pcDNA3.1 plasmids carrying wild-type CYP2C9*1, variant CYP2C9*3, and CYP2C9*13 cDNA. lornoxicam 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 15764717-3 2005 Formation of M4 was mediated specifically by CYP2C9 in human liver microsomes, as evidenced by the following observations: 1) cDNA-expressed CYP2C9-catalyzing formation of M4; 2) inhibition of M4 formation by sulfaphenazole, a CYP2C9-selective inhibitor; and 3) strong correlation between the production of M4 and CYP2C9-mediated tolbutamide 4-hydroxylase activities in a panel of human liver microsome samples. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15764717-3 2005 Formation of M4 was mediated specifically by CYP2C9 in human liver microsomes, as evidenced by the following observations: 1) cDNA-expressed CYP2C9-catalyzing formation of M4; 2) inhibition of M4 formation by sulfaphenazole, a CYP2C9-selective inhibitor; and 3) strong correlation between the production of M4 and CYP2C9-mediated tolbutamide 4-hydroxylase activities in a panel of human liver microsome samples. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15764717-3 2005 Formation of M4 was mediated specifically by CYP2C9 in human liver microsomes, as evidenced by the following observations: 1) cDNA-expressed CYP2C9-catalyzing formation of M4; 2) inhibition of M4 formation by sulfaphenazole, a CYP2C9-selective inhibitor; and 3) strong correlation between the production of M4 and CYP2C9-mediated tolbutamide 4-hydroxylase activities in a panel of human liver microsome samples. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15764711-8 2005 In a subsequent clinical study, the AUC of lornoxicam was increased by 1.9-fold and its oral clearance (CL/F) decreased by 44% in three CYP2C9*1/*13 subjects, compared with CYP2C9*1/*1 individuals. lornoxicam 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 15764717-7 2005 This mechanism is consistent with the structural modeling of the CYP2C9-diclofenac complex, which reveals that both the C-3" and C-4" of the dichlorophenyl ring are proximate to the heme group. Heme 182-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 15875171-0 2005 Twenty-four hour tolbutamide plasma concentration as a phenotypic measure of CYP2C9 activity. Tolbutamide 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 15764717-3 2005 Formation of M4 was mediated specifically by CYP2C9 in human liver microsomes, as evidenced by the following observations: 1) cDNA-expressed CYP2C9-catalyzing formation of M4; 2) inhibition of M4 formation by sulfaphenazole, a CYP2C9-selective inhibitor; and 3) strong correlation between the production of M4 and CYP2C9-mediated tolbutamide 4-hydroxylase activities in a panel of human liver microsome samples. Sulfaphenazole 209-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15764717-7 2005 This mechanism is consistent with the structural modeling of the CYP2C9-diclofenac complex, which reveals that both the C-3" and C-4" of the dichlorophenyl ring are proximate to the heme group. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 16192110-6 2005 There was good correlation between the formation rates of 4-OH valsartan and diclofenac 4"-hydroxylase activities (representative CYP2C9 activity) of 11 individual microsomes (r = 0.889). 4-oh valsartan 58-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 16192110-10 2005 These results showed that CYP2C9 is the only form responsible for 4-hydroxylation of valsartan in human liver microsomes. Valsartan 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 16192110-11 2005 Although CYP2C9 is involved in valsartan metabolism, CYP-mediated drug-drug interaction between valsartan and other co-administered drugs would be negligible. Valsartan 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 15901588-8 2005 Rosuvastatin undergoes only minor metabolism (10% of the administered dose) by the cytochrome P-450 2C9 isoenzyme. Rosuvastatin Calcium 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-103 15857618-5 2005 A range of substrates was investigated to determine the effect of the heme-substrate interaction on CYP2C9 redox potential. Heme 70-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 15842554-0 2005 Chlorpropamide 2-hydroxylation is catalysed by CYP2C9 and CYP2C19 in vitro: chlorpropamide disposition is influenced by CYP2C9, but not by CYP2C19 genetic polymorphism. Chlorpropamide 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 15961979-2 2005 Because CYP2C9 is a genetically polymorphic enzyme, genetic variability could play an important role in the potential interaction between NSAIDs and coumarins. Coumarins 149-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 15961979-10 2005 The risk of overanticoagulation was 2.98 (95% confidence interval, 1.09-7.02) in coumarin-treated patients taking NSAIDs with a CYP2C9*2 allele and 10.8 (95% confidence interval, 2.57-34.6) in those with a CYP2C9*3 allele. coumarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 15961979-10 2005 The risk of overanticoagulation was 2.98 (95% confidence interval, 1.09-7.02) in coumarin-treated patients taking NSAIDs with a CYP2C9*2 allele and 10.8 (95% confidence interval, 2.57-34.6) in those with a CYP2C9*3 allele. coumarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 15842554-5 2005 In incubation studies using human recombinant CYP isoforms, however, 2-OH-chlorpropamide was formed by both CYP2C9 and CYP2C19 with similar intrinsic clearances (CYP2C9 vs. CYP2C19: 0.26 vs. 0.22 microl min(-1) nmol(-1) protein). CHEBI:143296 69-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 15842554-0 2005 Chlorpropamide 2-hydroxylation is catalysed by CYP2C9 and CYP2C19 in vitro: chlorpropamide disposition is influenced by CYP2C9, but not by CYP2C19 genetic polymorphism. Chlorpropamide 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 15842554-0 2005 Chlorpropamide 2-hydroxylation is catalysed by CYP2C9 and CYP2C19 in vitro: chlorpropamide disposition is influenced by CYP2C9, but not by CYP2C19 genetic polymorphism. Chlorpropamide 76-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 15842554-7 2005 In in vivo clinical trials, eight subjects with the CYP2C9*1/*3 genotype exhibited significantly lower nonrenal clearance [*1/*3 vs.*1/*1: 1.8 +/- 0.2 vs. 2.4 +/- 0.1 ml h(-1) kg(-1), P < 0.05; 95% confidence interval (CI) on the difference 0.2, 1.0] and higher metabolic ratios (of chlorpropamide/2-OH-chlorpropamide in urine: *1/*3 vs.*1/*1: 1.01 +/- 0.19 vs. 0.56 +/- 0.08, P < 0.05; 95% CI on the difference - 0.9, - 0.1) than did 13 subjects with CYP2C9*1/*1 genotype. Chlorpropamide 286-300 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 15842554-7 2005 In in vivo clinical trials, eight subjects with the CYP2C9*1/*3 genotype exhibited significantly lower nonrenal clearance [*1/*3 vs.*1/*1: 1.8 +/- 0.2 vs. 2.4 +/- 0.1 ml h(-1) kg(-1), P < 0.05; 95% confidence interval (CI) on the difference 0.2, 1.0] and higher metabolic ratios (of chlorpropamide/2-OH-chlorpropamide in urine: *1/*3 vs.*1/*1: 1.01 +/- 0.19 vs. 0.56 +/- 0.08, P < 0.05; 95% CI on the difference - 0.9, - 0.1) than did 13 subjects with CYP2C9*1/*1 genotype. Chlorpropamide 286-300 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 458-464 15842554-0 2005 Chlorpropamide 2-hydroxylation is catalysed by CYP2C9 and CYP2C19 in vitro: chlorpropamide disposition is influenced by CYP2C9, but not by CYP2C19 genetic polymorphism. Chlorpropamide 76-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 15842554-7 2005 In in vivo clinical trials, eight subjects with the CYP2C9*1/*3 genotype exhibited significantly lower nonrenal clearance [*1/*3 vs.*1/*1: 1.8 +/- 0.2 vs. 2.4 +/- 0.1 ml h(-1) kg(-1), P < 0.05; 95% confidence interval (CI) on the difference 0.2, 1.0] and higher metabolic ratios (of chlorpropamide/2-OH-chlorpropamide in urine: *1/*3 vs.*1/*1: 1.01 +/- 0.19 vs. 0.56 +/- 0.08, P < 0.05; 95% CI on the difference - 0.9, - 0.1) than did 13 subjects with CYP2C9*1/*1 genotype. CHEBI:143296 301-320 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 15842554-3 2005 To evaluate whether genetic polymorphisms of CYP2C9 and/or CYP2C19 influence the disposition of chlorpropamide, a single oral dose of 250 mg chlorpropamide was administered to 21 healthy subjects pregenotyped for CYP2C9 and CYP2C19. Chlorpropamide 96-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15842554-7 2005 In in vivo clinical trials, eight subjects with the CYP2C9*1/*3 genotype exhibited significantly lower nonrenal clearance [*1/*3 vs.*1/*1: 1.8 +/- 0.2 vs. 2.4 +/- 0.1 ml h(-1) kg(-1), P < 0.05; 95% confidence interval (CI) on the difference 0.2, 1.0] and higher metabolic ratios (of chlorpropamide/2-OH-chlorpropamide in urine: *1/*3 vs.*1/*1: 1.01 +/- 0.19 vs. 0.56 +/- 0.08, P < 0.05; 95% CI on the difference - 0.9, - 0.1) than did 13 subjects with CYP2C9*1/*1 genotype. CHEBI:143296 301-320 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 458-464 15842554-9 2005 CONCLUSIONS: These results suggest that chlorpropamide disposition is principally determined by CYP2C9 activity in vivo, although both CYP2C9 and CYP2C19 have a catalysing activity of chlorpropamide 2-hydroxylation pathway. Chlorpropamide 40-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 15842554-9 2005 CONCLUSIONS: These results suggest that chlorpropamide disposition is principally determined by CYP2C9 activity in vivo, although both CYP2C9 and CYP2C19 have a catalysing activity of chlorpropamide 2-hydroxylation pathway. Chlorpropamide 184-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 15842554-5 2005 In incubation studies using human recombinant CYP isoforms, however, 2-OH-chlorpropamide was formed by both CYP2C9 and CYP2C19 with similar intrinsic clearances (CYP2C9 vs. CYP2C19: 0.26 vs. 0.22 microl min(-1) nmol(-1) protein). CHEBI:143296 69-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 15900281-0 2005 CYP2C9 haplotype structure in European American warfarin patients and association with clinical outcomes. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15900281-9 2005 CONCLUSION: These data establish a whole-gene, high-resolution haplotype structure for CYP2C9 in a European American patient population and suggest that genetic variation in exons, rather than the promoter or other regulatory regions, is largely responsible for warfarin sensitivity associated with CYP2C9 variants in this population. Warfarin 262-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15900282-1 2005 BACKGROUND: Warfarin dose response is partially explained by the polymorphisms in the cytochrome P450 (CYP) 2C9 gene, affecting S -warfarin clearance, as well as by age and body weight. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-111 15900282-1 2005 BACKGROUND: Warfarin dose response is partially explained by the polymorphisms in the cytochrome P450 (CYP) 2C9 gene, affecting S -warfarin clearance, as well as by age and body weight. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-111 15840038-2 2005 Recent studies suggest that an endothelial cytochrome P450 (CYP) epoxygenase (CYP 2C9) can modulate endothelium-dependent vasodilatation in two different ways: (1) by the production of epoxyeicosatrienoic acids (EETs), which elicit hyperpolarization and relaxation; and (2) by the release of oxygen-derived free radicals, which compromise the bioavailability of nitric oxide. epoxyeicosatrienoic acids 185-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 15840038-2 2005 Recent studies suggest that an endothelial cytochrome P450 (CYP) epoxygenase (CYP 2C9) can modulate endothelium-dependent vasodilatation in two different ways: (1) by the production of epoxyeicosatrienoic acids (EETs), which elicit hyperpolarization and relaxation; and (2) by the release of oxygen-derived free radicals, which compromise the bioavailability of nitric oxide. eets 212-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 15840038-2 2005 Recent studies suggest that an endothelial cytochrome P450 (CYP) epoxygenase (CYP 2C9) can modulate endothelium-dependent vasodilatation in two different ways: (1) by the production of epoxyeicosatrienoic acids (EETs), which elicit hyperpolarization and relaxation; and (2) by the release of oxygen-derived free radicals, which compromise the bioavailability of nitric oxide. oxygen-derived free radicals 292-320 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 15840038-2 2005 Recent studies suggest that an endothelial cytochrome P450 (CYP) epoxygenase (CYP 2C9) can modulate endothelium-dependent vasodilatation in two different ways: (1) by the production of epoxyeicosatrienoic acids (EETs), which elicit hyperpolarization and relaxation; and (2) by the release of oxygen-derived free radicals, which compromise the bioavailability of nitric oxide. Nitric Oxide 362-374 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 15840038-5 2005 In HD patients, ACh infusions were repeated together with sulfaphenazole (SPZ, 6 mg/min), a highly selective inhibitor of CYP 2C9 with and without concomitant blockade of the nitric oxide synthase (NOS) by N(omega)monomethyl L-arginine (L-NMMA, 16 microumol/min). Sulfaphenazole 58-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-129 16012074-3 2005 Recombinant human CYP3A4 and CYP2C9 both exhibited catalytic activity with respect to the formation of rotameric O-deethylated metabolites (M12, M13), the metabolites resulting from aromatization (M22/M24) and N-oxide reduction (M31). n-oxide 210-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 16012077-5 2005 When estazolam was incubated with expressed human CYP enzymes (CYP1A2, CYP2A6, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4), it was metabolized only by CYP3A4. Estazolam 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 15889670-0 2005 [Significance of cytochrome P450 2C9 genotype for the bleeding complications in patients treated with acenocoumarol]. Acenocoumarol 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-36 15766564-0 2005 Eicosapentaenoic acid metabolism by cytochrome P450 enzymes of the CYP2C subfamily. Eicosapentaenoic Acid 0-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-72 15766564-1 2005 CYP2C enzymes epoxidize arachidonic acid (AA) to metabolites involved in the regulation of vascular and renal function. Arachidonic Acid 24-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-5 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosapentaenoic Acid 31-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-69 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosapentaenoic Acid 31-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-134 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Fatty Acids, Unsaturated 99-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-69 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Fatty Acids, Unsaturated 99-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-134 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosanoids 182-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-69 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosanoids 182-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-134 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosapentaenoic Acid 206-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-69 15766564-7 2005 These results demonstrate that EPA is an efficient substrate of CYP2C enzymes and suggest that n-3 PUFA-rich diets may shift the CYP2C-dependent generation of physiologically active eicosanoids from AA- to EPA-derived metabolites. Eicosapentaenoic Acid 206-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-134 15889670-4 2005 AIM: The influence of CYP2C9 polymorphism on the effectiveness of the--in Hungary for oral anticoagulation exclusively used--acenocoumarol therapy and on the occurrence of bleeding complications was investigated. Acenocoumarol 125-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 15889670-8 2005 In the 145 patients bearing the alleles with reduced activity (CYP2C9*2 and/or *3), the optimised dose of the acenocoumarol was significantly (p < 0.001) lower than in patients with the wild type allele (2.12 +/- 0.96 mg/day and 2.90 +/- 1.45 mg/day, respectively). Acenocoumarol 110-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15805193-5 2005 Phenytoin is principally metabolized by CYP2C9, and both are probable substrates of the drug transporter P-glycoprotein. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15805193-7 2005 We report that a known functional polymorphism in CYP2C9 is highly associated with the maximum dose of phenytoin (P = 0.0066). Phenytoin 103-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 15801938-2 2005 Because gemfibrozil inhibits CYP2C9 in vitro, we studied its effects on the pharmacokinetics and pharmacodynamics of racemic warfarin. Gemfibrozil 8-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 15841315-0 2005 Prospective dosing of warfarin based on cytochrome P-450 2C9 genotype. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-60 15855721-1 2005 Two research groups have reported the effect of genetic polymorphisms of CYP2C9 and CYP2C19 on the pharmacokinetic parameters of phenytoin in Japanese epileptic patients. Phenytoin 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 15841315-1 2005 Cytochrome P-450 2C9 (CYP2C9) polymorphisms (CYP2C9*2 and CYP2C9*3) reduce the clearance of warfarin, increase the risk of bleeding, and prolong the time to stable dosing. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-20 15841315-1 2005 Cytochrome P-450 2C9 (CYP2C9) polymorphisms (CYP2C9*2 and CYP2C9*3) reduce the clearance of warfarin, increase the risk of bleeding, and prolong the time to stable dosing. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 15841315-1 2005 Cytochrome P-450 2C9 (CYP2C9) polymorphisms (CYP2C9*2 and CYP2C9*3) reduce the clearance of warfarin, increase the risk of bleeding, and prolong the time to stable dosing. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15841315-1 2005 Cytochrome P-450 2C9 (CYP2C9) polymorphisms (CYP2C9*2 and CYP2C9*3) reduce the clearance of warfarin, increase the risk of bleeding, and prolong the time to stable dosing. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15841315-3 2005 We initiated warfarin therapy in 48 orthopedic patients tailored to the following variables: CYP2C9 genotype, age, weight, height, gender, race, and use of simvastatin or amiodarone. Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15841315-4 2005 By using pharmacogenetics-based dosing, patients with a CYP2C9 variant achieved a stable, therapeutic warfarin dose without excessive delay. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 15608129-0 2005 Development and validation of a high-throughput radiometric cyp2c9 inhibition assay using tritiated diclofenac. tritiated diclofenac 90-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 15608129-3 2005 The assay is based on the release of tritium as tritiated water that occurs upon CYP2C9-mediated 4"-hydroxylation of diclofenac labeled with tritium in the 4" position. Tritium 37-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15608129-3 2005 The assay is based on the release of tritium as tritiated water that occurs upon CYP2C9-mediated 4"-hydroxylation of diclofenac labeled with tritium in the 4" position. Water 58-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15608129-3 2005 The assay is based on the release of tritium as tritiated water that occurs upon CYP2C9-mediated 4"-hydroxylation of diclofenac labeled with tritium in the 4" position. Diclofenac 117-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15608129-3 2005 The assay is based on the release of tritium as tritiated water that occurs upon CYP2C9-mediated 4"-hydroxylation of diclofenac labeled with tritium in the 4" position. Tritium 141-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15608129-5 2005 The reaction is NADPH-dependent, and sensitive to CYP2C9 inhibitors and inhibitory monoclonal antibodies, but not to inhibitors of or antibodies against other P450 enzymes. NADP 16-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 15608129-6 2005 Competition experiments using tritiated and unlabeled diclofenac indicated that CYP2C9-mediated diclofenac 4"-hydroxylation exhibits positive cooperativity and no significant kinetic isotope effect or NIH shift. Diclofenac 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15608129-6 2005 Competition experiments using tritiated and unlabeled diclofenac indicated that CYP2C9-mediated diclofenac 4"-hydroxylation exhibits positive cooperativity and no significant kinetic isotope effect or NIH shift. Diclofenac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15608129-9 2005 This assay thus represents a high-throughput version of the classic diclofenac 4"-hydroxylation assay, which is one of the most widely used methods to assess the potential for CYP2C9 inhibition of new chemical entities. diclofenac 4" 68-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 15703368-5 2005 Oxymorphone ER exhibits a minimal potential for causing metabolic drug-drug interactions mediated by CYP2C9 or CYP3A4. Oxymorphone 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 15660356-3 2005 The transgenic rice plant 2C9-57R2 expressing CYP2C9 gene showed resistance to sulfonylureas, and the transgenic rice plant 2C19-12R1 expressing CYP2C19 gene showed cross-resistance to certain herbicides with different structures and modes of action. Sulfonylurea Compounds 79-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 15855725-3 2005 This formation was completely blocked by both sulfaphenazole, a selective CYP2C9 inhibitor, and CYP2C9 antibody, whereas potent inhibitors selective for other CYPs exhibited little effects. Sulfaphenazole 46-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15855725-4 2005 Of 12 recombinant CYPs examined, O-desmethyl metabolite was principally formed by CYP2C9. o-desmethyl 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 15855726-7 2005 Phenytoin is metabolized to 4"-HPPH by CYP2C9 and CYP2C19 in which there are genetic polymorphisms. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 15855726-7 2005 Phenytoin is metabolized to 4"-HPPH by CYP2C9 and CYP2C19 in which there are genetic polymorphisms. hydroxyphenytoin 28-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 15676042-1 2005 AIMS: To investigate the potential induction by rifampicin of intestinal CYP2C8, CYP2C9, CYP2D6 and CYP3A4 using preparations of human enterocytes. Rifampin 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15652503-0 2005 Biosynthesis of epoxyeicosatrienoic acids varies between polymorphic CYP2C enzymes. epoxyeicosatrienoic acids 16-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-74 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. Arachidonic Acid 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. Arachidonic Acid 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-58 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. epoxyeicosatrienoic acids 63-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. epoxyeicosatrienoic acids 63-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-58 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. eets 90-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-58 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. 11,12-epoxy-5,8,14-eicosatrienoic acid 136-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-51 15652503-1 2005 Arachidonic acid is oxidized by cytochromes P450 2C (CYP2C) to epoxyeicosatrienoic acids (EETs), possessing vasoactive properties, with 11,12-EET as the endothelium derived hyperpolarization factor. 11,12-epoxy-5,8,14-eicosatrienoic acid 136-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-58 15652503-9 2005 In conclusion, there are genetic differences in the CYP2C-dependent oxidation of arachidonic acid to vasoactive metabolites, of which the relevance to cardiovascular pathophysiology is still unclear. Arachidonic Acid 81-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-57 15569819-5 2005 Overexpression of CYP2C9 increased COX-2 promoter activity, an effect accompanied by a significant increase in COX-2 protein expression and elevated prostacyclin production. Epoprostenol 149-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15569819-6 2005 The CYP2C9-induced expression of COX-2 was inhibited by the CYP2C9 inhibitor, sulfaphenazole, whereas 11,12-EET increased COX-2 expression. Sulfaphenazole 78-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 15569819-6 2005 The CYP2C9-induced expression of COX-2 was inhibited by the CYP2C9 inhibitor, sulfaphenazole, whereas 11,12-EET increased COX-2 expression. Sulfaphenazole 78-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 15569819-7 2005 Overexpression of CYP2C9 and stimulation with 11,12-EET increased intracellular cAMP levels and stimulated DNA-binding of the cAMP-response element-binding protein. Cyclic AMP 80-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15569819-7 2005 Overexpression of CYP2C9 and stimulation with 11,12-EET increased intracellular cAMP levels and stimulated DNA-binding of the cAMP-response element-binding protein. Cyclic AMP 126-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15569819-8 2005 The protein kinase A inhibitor, KT5720, attenuated the CYP2C9-induced increase in COX-2 promoter activity and protein expression. KT 5720 32-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 15569819-9 2005 Overexpression of CYP2C9 stimulated endothelial tube formation, an effect that was attenuated by the COX-2 inhibitor celecoxib. Celecoxib 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15569819-11 2005 CONCLUSIONS: These data indicate that CYP2C9-derived EETs induce the expression of COX-2 in endothelial cells via a cAMP-dependent pathway and that this mechanism contributes to CYP2C9-induced angiogenesis. Cyclic AMP 116-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 15569819-11 2005 CONCLUSIONS: These data indicate that CYP2C9-derived EETs induce the expression of COX-2 in endothelial cells via a cAMP-dependent pathway and that this mechanism contributes to CYP2C9-induced angiogenesis. Cyclic AMP 116-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 15569819-12 2005 Overexpression of cytochrome P450 (CYP) 2C9 in endothelial cells increased cAMP levels, stimulated the cAMP-response element-binding protein, and enhanced cyclooxygenase-2 (COX-2) promoter activity, protein expression, and prostacyclin production. Cyclic AMP 75-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-43 15569819-12 2005 Overexpression of cytochrome P450 (CYP) 2C9 in endothelial cells increased cAMP levels, stimulated the cAMP-response element-binding protein, and enhanced cyclooxygenase-2 (COX-2) promoter activity, protein expression, and prostacyclin production. Cyclic AMP 103-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-43 15569819-12 2005 Overexpression of cytochrome P450 (CYP) 2C9 in endothelial cells increased cAMP levels, stimulated the cAMP-response element-binding protein, and enhanced cyclooxygenase-2 (COX-2) promoter activity, protein expression, and prostacyclin production. Epoprostenol 223-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-43 15569819-13 2005 CYP2C9 overexpression stimulated endothelial tube formation, which was attenuated by the COX-2 inhibitor celecoxib. Celecoxib 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15676042-11 2005 Induction of intestinal CYP2C8 and CYP2C9 might contribute in part to rifampicin - mediated drug interactions, in addition to their hepatic counterparts and intestinal and hepatic CYP3A4. Rifampin 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 15537834-5 2005 R-Lansoprazole increased the Michaelis-Menten-derived V(max) of phenytoin 4-hydroxylation from 0.024 to 0.121 pmol/min/pmol P450, and lowered its K(m) from 20.5 to 15.0 microM, suggesting that R-lansoprazole activates CYP2C9-mediated phenytoin metabolism without displacing phenytoin from the active site. Dexlansoprazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15537834-0 2005 Lansoprazole enantiomer activates human liver microsomal CYP2C9 catalytic activity in a stereospecific and substrate-specific manner. Lansoprazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 15537834-1 2005 We recently proposed a possible stereoselective activation by lansoprazole of CYP2C9-catalyzed tolbutamide hydroxylation, as well as stereoselective inhibition of several cytochrome P450 (P450) isoforms. Lansoprazole 62-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15537834-1 2005 We recently proposed a possible stereoselective activation by lansoprazole of CYP2C9-catalyzed tolbutamide hydroxylation, as well as stereoselective inhibition of several cytochrome P450 (P450) isoforms. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15537834-2 2005 This study evaluated the effects of lansoprazole enantiomers on CYP2C9 activity in vitro, using several probe substrates. Lansoprazole 36-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 15537834-4 2005 R-Lansoprazole-mediated activation of the formation of 4-hydroxyphenytoin was also seen with recombinant human CYP2C9. Dexlansoprazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 15537834-4 2005 R-Lansoprazole-mediated activation of the formation of 4-hydroxyphenytoin was also seen with recombinant human CYP2C9. hydroxyphenytoin 55-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 15714076-0 2005 CYP2C9 gene variants, drug dose, and bleeding risk in warfarin-treated patients: a HuGEnet systematic review and meta-analysis. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15714076-1 2005 PURPOSE: Two common variant alleles of the cytochrome CYP2C9 (CYP2C9*2 and CYP2C9*3) lead to reduced warfarin metabolism in vitro and in vivo. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 15714076-1 2005 PURPOSE: Two common variant alleles of the cytochrome CYP2C9 (CYP2C9*2 and CYP2C9*3) lead to reduced warfarin metabolism in vitro and in vivo. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15714076-1 2005 PURPOSE: Two common variant alleles of the cytochrome CYP2C9 (CYP2C9*2 and CYP2C9*3) lead to reduced warfarin metabolism in vitro and in vivo. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15714076-2 2005 The study objective was to examine the strength and quality of existing evidence about CYP2C9 gene variants and clinical outcomes in warfarin-treated patients. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15714076-11 2005 Mean difference in daily warfarin dose: for CYP2C9*2, the reduction was 0.85 mg (0.60-1.11 mg), a 17% reduction. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 15714076-16 2005 CONCLUSIONS: Patients with CYP2C9*2 and CYP2C9*3 alleles have lower mean daily warfarin doses and a greater risk of bleeding. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 15714076-16 2005 CONCLUSIONS: Patients with CYP2C9*2 and CYP2C9*3 alleles have lower mean daily warfarin doses and a greater risk of bleeding. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15854203-3 2005 Metabolism of pitavastatin by the cytochrome P450 (CYP) system is minimal, principally through CYP 2C9, with little involvement of the CYP 3A4 isoenzyme, potentially reducing the risk of drug-drug interactions between pitavastatin and other drugs known to inhibit CYP enzymes. pitavastatin 14-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-102 15537834-5 2005 R-Lansoprazole increased the Michaelis-Menten-derived V(max) of phenytoin 4-hydroxylation from 0.024 to 0.121 pmol/min/pmol P450, and lowered its K(m) from 20.5 to 15.0 microM, suggesting that R-lansoprazole activates CYP2C9-mediated phenytoin metabolism without displacing phenytoin from the active site. michaelis-menten 29-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15537834-5 2005 R-Lansoprazole increased the Michaelis-Menten-derived V(max) of phenytoin 4-hydroxylation from 0.024 to 0.121 pmol/min/pmol P450, and lowered its K(m) from 20.5 to 15.0 microM, suggesting that R-lansoprazole activates CYP2C9-mediated phenytoin metabolism without displacing phenytoin from the active site. Phenytoin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15537834-10 2005 Overall, these results suggest that R-lansoprazole activates CYP2C9 in a stereospecific and substrate-specific manner, possibly by binding within the active site and inducing positive cooperativity. Dexlansoprazole 36-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 15547048-3 2005 Using P450 isoform-selective inhibitors and recombinant heterologously expressed enzymes, it was demonstrated that several P450 enzymes catalyzed sertraline N-demethylation, with CYP2B6 contributing the greatest extent, and lesser contributions from CYP2C19, CYP2C9, CYP3A4, and CYP2D6. sertraline n 146-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 259-265 15711768-0 2005 Retrospective analyses of acenocoumarol doses and bleeding complications in patients with wild type or variant cytochrome P450 CYP2C9 alleles. Acenocoumarol 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. Krypton 61-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. pyrrole-kr 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. o-demethylpyrrole-kr 91-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. Nitrogen 122-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. Krypton 80-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. Krypton 81-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15585365-6 2005 The results show that CYP3A4 contributes to the oxidation of KR-60436 to pyrrole-KR-60436, O-demethylpyrrole-KR-60436 and N-dehydroxyethyl-KR-60436, and CYP2C9 and CYP2D6 play roles in demethylation of KR-60436 to form the active metabolite, O-demethyl-KR-60436. o-demethyl-kr 242-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15675511-2 2005 In this approach, single-stranded DNA (ssDNA)-polyacrylamide (polyAAm) conjugate was used as a pseudo-immobilized affinity ligand to separate the target DNA, cytochrome P450 2C9 (CYP2C9), and its point mutant. polyacrylamide 46-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-177 15675511-2 2005 In this approach, single-stranded DNA (ssDNA)-polyacrylamide (polyAAm) conjugate was used as a pseudo-immobilized affinity ligand to separate the target DNA, cytochrome P450 2C9 (CYP2C9), and its point mutant. polyacrylamide 46-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15675511-2 2005 In this approach, single-stranded DNA (ssDNA)-polyacrylamide (polyAAm) conjugate was used as a pseudo-immobilized affinity ligand to separate the target DNA, cytochrome P450 2C9 (CYP2C9), and its point mutant. polyacrylamide 62-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-177 15675511-2 2005 In this approach, single-stranded DNA (ssDNA)-polyacrylamide (polyAAm) conjugate was used as a pseudo-immobilized affinity ligand to separate the target DNA, cytochrome P450 2C9 (CYP2C9), and its point mutant. polyacrylamide 62-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 15665498-2 2005 The modified CDs inhibited the activities of CYP2C19 and CYP3A4 while enhancing CYP2C9 activity by 140 to 176% relative to the control values at lower concentrations. Cyclodextrins 13-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15606435-0 2005 Lornoxicam pharmacokinetics in relation to cytochrome P450 2C9 genotype. lornoxicam 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 15606435-1 2005 AIMS: To investigate the pharmacokinetics of lornoxicam and the relationship with CYP2C9 polymorphism in healthy Chinese subjects. lornoxicam 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 15606435-5 2005 RESULTS: Of the 18 subjects, one subject was found to be a very poor metabolizer of lornoxicam with a long t(1/2) of 106 h, a low CL/F of 0.71 ml min(-1), and a high AUC(0-infinity) of 187.6 microg ml(-1) h. Genotyping studies revealed that this subject was heterozygous for CYP2C9*3 and a new variant CYP2C9 allele. lornoxicam 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 275-281 15606435-5 2005 RESULTS: Of the 18 subjects, one subject was found to be a very poor metabolizer of lornoxicam with a long t(1/2) of 106 h, a low CL/F of 0.71 ml min(-1), and a high AUC(0-infinity) of 187.6 microg ml(-1) h. Genotyping studies revealed that this subject was heterozygous for CYP2C9*3 and a new variant CYP2C9 allele. lornoxicam 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 302-308 15606435-8 2005 CONCLUSIONS: The results show that the pharmacokinetics of lornoxicam are dependent on CYP2C9 polymorphism. lornoxicam 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15606435-9 2005 In particular, the presence of the CYP2C9*3 allele impairs the oral clearance of lornoxicam. lornoxicam 81-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 15637526-8 2005 In addition to its role in pharmacokinetics, CYP2C9 contributes to the metabolism of fatty acids, prostanoids, and steroid hormones, and it may catalyze potentially toxic bioactivation reactions. Fatty Acids 85-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15637526-8 2005 In addition to its role in pharmacokinetics, CYP2C9 contributes to the metabolism of fatty acids, prostanoids, and steroid hormones, and it may catalyze potentially toxic bioactivation reactions. Prostaglandins 98-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15637526-8 2005 In addition to its role in pharmacokinetics, CYP2C9 contributes to the metabolism of fatty acids, prostanoids, and steroid hormones, and it may catalyze potentially toxic bioactivation reactions. Steroids 115-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. Sulfonylurea Compounds 36-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. Tolbutamide 52-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. Glyburide 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. Glyburide 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. glimepiride 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-6 2005 Total oral clearance of all studied sulphonylureas (tolbutamide, glibenclamide [glyburide], glimepiride, glipizide) was only about 20% in persons with the CYP2C9*3/*3 genotype compared with carriers of the wild-type genotype CYP2C9*1/*1, and clearance in the heterozygous carriers was between 50% and 80% of that of the wild-type genotypes. Glipizide 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 16372821-10 2005 The meglitinide-class drug nateglinide is metabolised by CYP2C9. meglitinide 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16372821-10 2005 The meglitinide-class drug nateglinide is metabolised by CYP2C9. Nateglinide 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 16372821-11 2005 According to the pharmacokinetic data, moderate dose adjustments based on CYP2C9 genotypes may help in reducing interindividual variability in the antihyperglycaemic effects of nateglinide. Nateglinide 177-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 16372821-14 2005 CYP2C8 and CYP3A4 are the main enzymes catalysing biotransformation of the thiazolidinediones troglitazone and pioglitazone, whereas rosiglitazone is metabolised by CYP2C9 and CYP2C8. Rosiglitazone 133-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 16372822-8 2005 More recently, CYP2C9 has also been reported to catalyse the hydroxylation of phenprocoumon and acenocoumarol. Phenprocoumon 78-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16372822-8 2005 More recently, CYP2C9 has also been reported to catalyse the hydroxylation of phenprocoumon and acenocoumarol. Acenocoumarol 96-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 16372822-10 2005 Overall, the CYP2C9 isoenzyme appears to be most important for the clearance of warfarin, followed by acenocoumarol and, lastly, phenprocoumon. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 16372822-10 2005 Overall, the CYP2C9 isoenzyme appears to be most important for the clearance of warfarin, followed by acenocoumarol and, lastly, phenprocoumon. Acenocoumarol 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15486075-0 2005 Interactions between CYP2C9 and CYP2C19 in reconstituted binary systems influence their catalytic activity: possible rationale for the inability of CYP2C19 to catalyze methoxychlor demethylation in human liver microsomes. Methoxychlor 168-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15486075-5 2005 When reconstituted with CPR, cytochrome b(5), and lipid, purified CYP2C19 and CYP2C9 catalyzed methoxychlor-O-demethylation. methoxychlor-o 95-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15486075-6 2005 However, whereas equimolar CPR to CYP2C9 supported maximal rates of methoxychlor demethylation and diclofenac hydroxylation, the rate of methoxychlor demethylation by CYP2C19 was not fully saturated, even with a 9-fold molar excess of CPR over CYP2C19. Methoxychlor 68-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 15486075-6 2005 However, whereas equimolar CPR to CYP2C9 supported maximal rates of methoxychlor demethylation and diclofenac hydroxylation, the rate of methoxychlor demethylation by CYP2C19 was not fully saturated, even with a 9-fold molar excess of CPR over CYP2C19. Diclofenac 99-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 15486075-10 2005 By contrast, in the incubation containing CYP2C9, diclofenac hydroxylation was activated by the presence of CYP2C19. Diclofenac 50-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 15617742-0 2005 The frequency of cytochrome P450 2C9 genetic variants in the Russian population and their associations with individual sensitivity to warfarin therapy. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-36 15940194-0 2005 [Clinical value of allele variants of cytochrome CYP2C9 gene for anticoagulation therapy with warfarin]. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 15940194-1 2005 Warfarin is metabolized by cytochrome CYP2C9 and its pharmacokinetic properties depend on structural polymorphisms of CYP2C9 gene. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 15940194-1 2005 Warfarin is metabolized by cytochrome CYP2C9 and its pharmacokinetic properties depend on structural polymorphisms of CYP2C9 gene. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 15940194-2 2005 We studied frequencies of allele variants of CYP2C9 gene and associations of individual reaction to warfarin intake with genotype of CYP2C9 gene. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 15940194-4 2005 Carriers of CYP2C9x2 and CYP2C9x3 alleles more rapidly achieved therapeutic levels of hypocoagulation and required significantly lower weekly doses of warfarin. Warfarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 15824753-0 2005 Influence of CYP2C9 polymorphisms, demographic factors and concomitant drug therapy on warfarin metabolism and maintenance dose. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15824753-2 2005 S-warfarin is metabolized mainly by polymorphic cytochrome P450 (CYP) 2C9. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-73 15824753-3 2005 We systematically quantified the influence of CYP2C9 genotype, demographic factors and concomitant drug treatment on warfarin metabolism and maintenance dose. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 15824753-5 2005 Multiple regression analysis demonstrated that CYP2C9 genotype, age, concomitant treatment with warfarin metabolism inducers and lean body weight contributed significantly to interindividual variability in warfarin dose requirement (adjusted R(2)=0.37). Warfarin 206-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 15578879-0 2004 Effect of polymorphisms in the cytochrome P450 CYP2C9 gene on warfarin anticoagulation. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 15578879-3 2004 Warfarin metabolism is under genetic control, involving primarily the CYP2C9 gene encoding the enzyme that catalyzes the conversion of warfarin to inactive metabolites. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 15578879-3 2004 Warfarin metabolism is under genetic control, involving primarily the CYP2C9 gene encoding the enzyme that catalyzes the conversion of warfarin to inactive metabolites. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 15578879-5 2004 The objective of this case study is to investigate the relationship between CYP2C9 genotype and warfarin anticoagulation. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 15578879-6 2004 DESIGN: A case of deep vein thrombosis treated with the standard warfarin dose is investigated for intensity of anticoagulation and CYP2C9 genotype; the case illustrates the relationship between CYP2C9 variant and overanticoagulation with subsequent bleeding complication. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 15578879-6 2004 DESIGN: A case of deep vein thrombosis treated with the standard warfarin dose is investigated for intensity of anticoagulation and CYP2C9 genotype; the case illustrates the relationship between CYP2C9 variant and overanticoagulation with subsequent bleeding complication. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 15578879-7 2004 RESULTS: The patient"s genotype, CYP2C9*1*3, correlated with an exaggerated anticoagulant response during the initiation of warfarin therapy at standard dose, and a bleeding episode ensued. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15578879-10 2004 Genotype knowledge of the CYP2C9 variant alleles may help the clinician to individualize warfarin therapy with the ultimate goals of shortening the initial period of induction therapy, reaching a stable maintenance dose earlier, and minimizing bleeding complications in patients who are high responders and need lower warfarin doses. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 15578879-10 2004 Genotype knowledge of the CYP2C9 variant alleles may help the clinician to individualize warfarin therapy with the ultimate goals of shortening the initial period of induction therapy, reaching a stable maintenance dose earlier, and minimizing bleeding complications in patients who are high responders and need lower warfarin doses. Warfarin 318-326 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 15333513-8 2004 Avasimibe inhibited CYP2C9 (IC50 2.9 microM), CYP1A2 (IC50 13.9 microM), and CYP2C19 (IC50 26.5 microM). avasimibe 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 15333513-9 2004 A clinical drug interaction study was conducted to determine whether avasimibe might interact with the CYP2C9 substrate warfarin. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 15342469-8 2004 Gomisin C, however, inhibited CYP1A2-, CYP2C9-, CYP2C19-, and CYP2D6-dependent activities only to a limited extent (IC50 values >10 microM). schizandrer A 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 15592327-0 2004 CYP2C9 polymorphisms and the interindividual variability in pharmacokinetics and pharmacodynamics of the loop diuretic drug torsemide. Torsemide 124-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. Torsemide 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-148 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. Torsemide 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. inn 53-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-148 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. inn 53-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. Torsemide 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-148 15592327-1 2004 INTRODUCTION: According to in vitro data, torsemide (INN, torasemide) is a substrate of the genetically polymorphic enzyme cytochrome P450 (CYP) 2C9, but the impact of CYP2C9 polymorphisms on torsemide pharmacokinetics and pharmacodynamics has not been studied in humans. Torsemide 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 15592327-3 2004 The effects of the CYP2C9 polymorphism on torsemide-induced urine volume and urinary elimination of sodium, potassium, chloride, and uric acid were measured during a salt-restricted diet. Torsemide 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 15592327-4 2004 RESULTS: Median torsemide total oral clearance values were 3.4, 2.2, and 1.2 L/h in carriers of the CYP2C9 genotypes *1/*1 , *1/*3 , and *3/*3 , respectively, but there was no significant difference related to CYP2C9*2 . Torsemide 16-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 15592327-6 2004 From 0 to 8 hours after torsemide administration, Na + , K + , and Cl - elimination was higher in carriers of CYP2C9*3 alleles than in carriers of the homozygous wild-type genotype, and 24-hour uric acid elimination values in urine were 451, 350, and 249 mg in carriers of 0, 1, and 2 CYP2C9*3 alleles, respectively (P = .003). Torsemide 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 15592327-7 2004 CONCLUSION: Torsemide pharmacokinetics differed significantly between subgroups with different CYP2C9 genotypes, and diuretic effects were slightly more exaggerated in carriers of CYP2C9*3 alleles. Torsemide 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 15592327-7 2004 CONCLUSION: Torsemide pharmacokinetics differed significantly between subgroups with different CYP2C9 genotypes, and diuretic effects were slightly more exaggerated in carriers of CYP2C9*3 alleles. Torsemide 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 15592335-2 2004 METHODS: The frequency of coadministration between losartan and well-established cytochrome P450 (CYP) 2C9 inhibitors, as well as codeine and tramadol and CYP2D6 inhibitors, was studied by use of data from a university hospital medication database. Losartan 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-106 15608560-0 2004 Upstream and coding region CYP2C9 polymorphisms: correlation with warfarin dose and metabolism. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 15585441-12 2004 As with other protease inhibitors, atazanavir is also a substrate and moderate inhibitor of the cytochrome P450 (CYP) system, in particular CYP3A4 and CYP2C9. Atazanavir Sulfate 35-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 15608560-1 2004 OBJECTIVES: To assess whether CYP2C9 alleles other than CYP2C9*2 and *3 are associated with a low-warfarin dose requirement and the relevance of upstream CYP2C9 polymorphisms to dose requirement and metabolism. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15608560-3 2004 PCR-based genotyping assays for novel upstream and other known polymorphisms were used to screen a larger patient population of known CYP2C9*2 and *3 genotype requiring a range of warfarin doses. Warfarin 180-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 15608560-10 2004 CONCLUSIONS: The coding region non-synonymous polymorphisms associated with the CYP2C9*2 and CYP2C9*3 alleles are the major CYP2C9-related factor affecting warfarin dose in UK Caucasians. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15608560-10 2004 CONCLUSIONS: The coding region non-synonymous polymorphisms associated with the CYP2C9*2 and CYP2C9*3 alleles are the major CYP2C9-related factor affecting warfarin dose in UK Caucasians. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15608560-10 2004 CONCLUSIONS: The coding region non-synonymous polymorphisms associated with the CYP2C9*2 and CYP2C9*3 alleles are the major CYP2C9-related factor affecting warfarin dose in UK Caucasians. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15536456-11 2004 Carriers of the CYP2C9*2 or *3 allele needed a significantly lower phenprocoumon dosage compared with homozygous CYP2C9*1/*1 subjects. Phenprocoumon 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 29793229-5 2004 CYP2C9 metabolizes several oral hypoglycemics, oral anticoagulants, non-steroidal anti-inflammatory drugs and other drugs, including phenytoin, losartan, fluvastatin, and torsemide. Phenytoin 133-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29793229-5 2004 CYP2C9 metabolizes several oral hypoglycemics, oral anticoagulants, non-steroidal anti-inflammatory drugs and other drugs, including phenytoin, losartan, fluvastatin, and torsemide. Losartan 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29793229-5 2004 CYP2C9 metabolizes several oral hypoglycemics, oral anticoagulants, non-steroidal anti-inflammatory drugs and other drugs, including phenytoin, losartan, fluvastatin, and torsemide. Fluvastatin 154-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29793229-5 2004 CYP2C9 metabolizes several oral hypoglycemics, oral anticoagulants, non-steroidal anti-inflammatory drugs and other drugs, including phenytoin, losartan, fluvastatin, and torsemide. Torsemide 171-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29793229-8 2004 Several studies demonstrated that the CYP2C9 polymorphisms are medically important for non-steroidal anti-inflammatory drugs, for oral hypoglycemics, vitamin K antagonistic oral anticoagulants, and phenytoin. Vitamin K 150-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 29793229-8 2004 Several studies demonstrated that the CYP2C9 polymorphisms are medically important for non-steroidal anti-inflammatory drugs, for oral hypoglycemics, vitamin K antagonistic oral anticoagulants, and phenytoin. Phenytoin 198-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 29793229-11 2004 Data appear mature enough for the routine consideration of CYP2C9 genotypes in therapy with acenocoumarol, phenytoin, warfarin, and some other drugs. Acenocoumarol 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 29793229-11 2004 Data appear mature enough for the routine consideration of CYP2C9 genotypes in therapy with acenocoumarol, phenytoin, warfarin, and some other drugs. Phenytoin 107-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 29793229-11 2004 Data appear mature enough for the routine consideration of CYP2C9 genotypes in therapy with acenocoumarol, phenytoin, warfarin, and some other drugs. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 15557548-8 2004 Phenytoin and phenobarbital are metabolized by cytochrome P450 isozymes, with activity dependent on genetic polymorphism (CYP2C9, CYP2C19). Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 15557548-8 2004 Phenytoin and phenobarbital are metabolized by cytochrome P450 isozymes, with activity dependent on genetic polymorphism (CYP2C9, CYP2C19). Phenobarbital 14-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 15543094-9 2004 Chronic hypoxia, increased amounts of nonheme iron in the liver and adrenals of these infants, enhanced activity of CYP2C9 regarded as the functional source of reactive oxygen species (ROS) in some endothelial cells, and nicotine accumulation in tissues also intensified production of ROS. Reactive Oxygen Species 160-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 15543094-9 2004 Chronic hypoxia, increased amounts of nonheme iron in the liver and adrenals of these infants, enhanced activity of CYP2C9 regarded as the functional source of reactive oxygen species (ROS) in some endothelial cells, and nicotine accumulation in tissues also intensified production of ROS. Reactive Oxygen Species 185-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 15543094-9 2004 Chronic hypoxia, increased amounts of nonheme iron in the liver and adrenals of these infants, enhanced activity of CYP2C9 regarded as the functional source of reactive oxygen species (ROS) in some endothelial cells, and nicotine accumulation in tissues also intensified production of ROS. Reactive Oxygen Species 285-288 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 15304426-0 2004 The involvement of CYP3A4 and CYP2C9 in the metabolism of 17 alpha-ethinylestradiol. Ethinyl Estradiol 58-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15536456-13 2004 Similar to warfarin and acenocoumarol, phenprocoumon had a lower dosage requirement in carriers of CYP2C9*2 or *3 compared with that in CYP2C9 wild-type subjects. Phenprocoumon 39-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 15304426-6 2004 The involvement of CYP3A4 and CYP2C9 was further confirmed by using selective chemical inhibitors (i.e., ketoconazole and sulfaphenazole). Ketoconazole 105-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15304426-6 2004 The involvement of CYP3A4 and CYP2C9 was further confirmed by using selective chemical inhibitors (i.e., ketoconazole and sulfaphenazole). Sulfaphenazole 122-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15304427-10 2004 Aprepitant was a very weak inhibitor of CYP2C9 and CYP2C19, with Ki values of 108 and 66 microM for the 7-hydroxylation of warfarin and the 4"-hydroxylation of S-mephenytoin, respectively. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15373932-2 2004 Rosiglitazone is a novel thiazolidinedione antidiabetic drug, mainly metabolized by CYP2C8 and to a lesser extent CYP2C9. Rosiglitazone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 15304427-10 2004 Aprepitant was a very weak inhibitor of CYP2C9 and CYP2C19, with Ki values of 108 and 66 microM for the 7-hydroxylation of warfarin and the 4"-hydroxylation of S-mephenytoin, respectively. Mephenytoin 160-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15528841-5 2004 In contrast, in addition to the reported inhibitory effect of isoniazid on CYP1A2, CYP2A6, CYP2C19, and CYP3A activities, our results newly showed its effect on CYP2C9 and CYP2E1 activities. Isoniazid 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 15569425-0 2004 [Association between genetic polymorphisms of CYP2C19 and CYP2C9 and phenytoin serum concentration]. Phenytoin 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15569425-1 2004 OBJECTIVE: To investigate the relationship between the genetic polymorphism of cytochrome CYP2C19 and CYP2C9 and the serum concentration of phenytoin (PHT) in patients with epilepsy. Phenytoin 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 15569425-1 2004 OBJECTIVE: To investigate the relationship between the genetic polymorphism of cytochrome CYP2C19 and CYP2C9 and the serum concentration of phenytoin (PHT) in patients with epilepsy. Phenytoin 151-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 15569425-11 2004 CONCLUSION: Phenytoin is metabolized via CYP2C19 and CYP2C9. Phenytoin 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 15373932-2 2004 Rosiglitazone is a novel thiazolidinedione antidiabetic drug, mainly metabolized by CYP2C8 and to a lesser extent CYP2C9. 2,4-thiazolidinedione 25-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 15373932-9 2004 CONCLUSIONS: This study revealed that ketoconazole affected the disposition of rosiglitazone in humans, probably by the inhibition of CYP2C8 and CYP2C9, leading to increasing rosiglitazone concentrations that could increase the efficacy of rosiglitazone or its adverse events. Ketoconazole 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 15373932-9 2004 CONCLUSIONS: This study revealed that ketoconazole affected the disposition of rosiglitazone in humans, probably by the inhibition of CYP2C8 and CYP2C9, leading to increasing rosiglitazone concentrations that could increase the efficacy of rosiglitazone or its adverse events. Rosiglitazone 79-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 15373932-9 2004 CONCLUSIONS: This study revealed that ketoconazole affected the disposition of rosiglitazone in humans, probably by the inhibition of CYP2C8 and CYP2C9, leading to increasing rosiglitazone concentrations that could increase the efficacy of rosiglitazone or its adverse events. Rosiglitazone 175-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 15373932-9 2004 CONCLUSIONS: This study revealed that ketoconazole affected the disposition of rosiglitazone in humans, probably by the inhibition of CYP2C8 and CYP2C9, leading to increasing rosiglitazone concentrations that could increase the efficacy of rosiglitazone or its adverse events. Rosiglitazone 175-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Phenobarbital 61-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 15382119-4 2004 We show that IL-1beta decreases CAR expression and decreases phenobarbital- or bilirubin-mediated induction of CYP2B6, CYP2C9, CYP3A4, UGT1A1, GSTA1, GSTA2, and SLC21A6 messenger RNA. Bilirubin 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 15540908-0 2004 Genetic polymorphism of the CYP2C9 subfamily of 3 different races in warfarin maintenance dose. Warfarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 15454736-0 2004 A novel CYP2C9 variant that caused erroneous genotyping in a patient on warfarin therapy. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 15385837-0 2004 Dosage recommendation of phenytoin for patients with epilepsy with different CYP2C9/CYP2C19 polymorphisms. Phenytoin 25-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 15325841-1 2004 The effect of human serum albumin (HSA), in its endogenous, free fatty acid free (FAF) and globulin free (GF) form, on the activity of CYP2C9 was studied in human liver microsomes using tolbutamide as the substrate. Fatty Acids, Nonesterified 60-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 15325841-1 2004 The effect of human serum albumin (HSA), in its endogenous, free fatty acid free (FAF) and globulin free (GF) form, on the activity of CYP2C9 was studied in human liver microsomes using tolbutamide as the substrate. Fatty Acids, Nonesterified 82-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 15325841-1 2004 The effect of human serum albumin (HSA), in its endogenous, free fatty acid free (FAF) and globulin free (GF) form, on the activity of CYP2C9 was studied in human liver microsomes using tolbutamide as the substrate. Tolbutamide 186-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 15325841-4 2004 HSA(FAF) and BSA showed a concentration-dependent and biphasic (activation and inhibition) interaction with CYP2C9 activity. Fatty Acids, Nonesterified 4-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 15325841-10 2004 The FDH Test ratio for HSA was about 1, indicating that HSA binding of tolbutamide reduced the CYP2C9 activity in accord with the free-drug hypothesis. Tolbutamide 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 15325841-13 2004 As revealed by their removal, free fatty acids and globulins, significantly alter the interaction of HSA with CYP2C9. Fatty Acids, Nonesterified 30-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 15325841-14 2004 In addition, HSA and BSA showed different effects on the oxidation of tolbutamide by CYP2C9. Tolbutamide 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. beta-Naphthoflavone 50-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15364537-8 2004 Primary cultures of hepatocytes were treated with beta-naphthoflavone, an inducer of CYP1A2, phenobarbital or rifampin, both of which induce CYP2B6, CYP2C9, CYP2C19, and CYP3A4, albeit it to different extents. Rifampin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15288804-0 2004 Active-site characteristics of CYP2C19 and CYP2C9 probed with hydantoin and barbiturate inhibitors. Hydantoins 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 15288804-0 2004 Active-site characteristics of CYP2C19 and CYP2C9 probed with hydantoin and barbiturate inhibitors. barbituric acid 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 15288804-5 2004 (S)-(+)-N-3-Benzylnirvanol and (R)-(-)-N-3-benzylphenobarbital emerged as the most potent and selective CYP2C19 inhibitors, with K(i) values of < 250nM--at least two orders of magnitude greater inhibitor potency than towards CYP2C9. N-3-benzylnirvanol 0-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 228-234 15288804-5 2004 (S)-(+)-N-3-Benzylnirvanol and (R)-(-)-N-3-benzylphenobarbital emerged as the most potent and selective CYP2C19 inhibitors, with K(i) values of < 250nM--at least two orders of magnitude greater inhibitor potency than towards CYP2C9. (r)-(-)-n-3-benzylphenobarbital 31-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 228-234 15371982-0 2004 Novel CYP2C9 genetic variants in Asian subjects and their influence on maintenance warfarin dose. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 15371982-1 2004 BACKGROUND: Commonly occurring genetic variants in CYP2C9 are known to reduce catalytic activity and are associated with enhanced patient sensitivity to warfarin. Warfarin 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 15371982-2 2004 Interethnic differences in warfarin dose requirement have been described in the Asian population, and we postulate that this could be related to genetic variants of CYP2C9 that are unique to ethnic groups. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 15371982-3 2004 METHODS: We prospectively genotyped 125 patients who were receiving a stable daily warfarin dose to maintain international normalized ratio values between 2 and 3 through comprehensive sequencing of the promoter and coding regions of the CYP2C9 gene. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 238-244 15371982-7 2004 CYP2C9*3, but not CYP2C9*2, was found in Chinese and Malay patients, and carriers of the CYP2C9*3 variant in Chinese ( P <.01) and Indian ( P <.01) patients, but not Malay patients ( P =.77), required less warfarin. Warfarin 206-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15341704-1 2004 Warfarin, the principal oral anticoagulant used in the treatment and prevention of thromboembolic disease, is primarily metabolized by CYP2C9, one of the hepatic microsomal enzymes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 15319344-5 2004 Other metabolites formed in NADPH-fortified liver microsomes included the TZD-5-OH derivative (M24), also catalyzed by CYP3A4, and the O-desmethyl derivative (M28), whose formation was catalyzed by CYP2C9 and CYP2C19. NADP 28-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 15350166-6 2004 The demonstration of the safe use of fluvastatin in a wide range of patients may be associated with the predominant acid form of the drug in vivo, as well as its predominant metabolism via the cytochrome P450 2C9 pathway. Fluvastatin 37-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-212 15378224-2 2004 METHODS: The inhibitory potency of memantine on CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 activities was examined with specific probe drugs in HLM and recombinant P450s. Memantine 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 15295798-2 2004 Torasemide was reported to be mainly metabolized via hepatic CYP2C9 in humans, and human CYP2C9 and male rat CYP2C11 proteins have 77% homology. Torsemide 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 15742978-0 2004 Genetic polymorphisms of cytochrome P450 2C9 causing reduced phenprocoumon (S)-7-hydroxylation in vitro and in vivo. ethyl-2-methylthio-4-methyl-5-pyrimidine carboxylate 75-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-44 15351856-1 2004 Variant cytochrome P450 (CYP) 2C9 genotypes are associated with low maintenance dose requirement of warfarin therapy and increased risk of major bleeding events. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-33 15742978-1 2004 The effect of cytochrome P450 (CYP) 2C9 polymorphisms on the stereoselective biotransformation of the oral anticoagulant phenprocoumon (PPC) to inactive, monohydroxylated metabolites was studied in vitro and in vivo. Phenprocoumon 121-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-39 15742978-2 2004 In human liver microsomes, the (S)-7-hydroxylation--being the major metabolic pathway--was significantly compromised in a gene-dose-dependent manner in samples expressing the CYP2C9*2 or CYP2C9*3 allele. Sulfur 32-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 15742978-2 2004 In human liver microsomes, the (S)-7-hydroxylation--being the major metabolic pathway--was significantly compromised in a gene-dose-dependent manner in samples expressing the CYP2C9*2 or CYP2C9*3 allele. Sulfur 32-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 15742978-3 2004 The CYP2C9*3/*3 genotype corresponded to an almost fourfold lower (S)-7-hydroxylation rate than CYP2C9*1/*1 (wild-type). Sulfur 67-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 15742978-4 2004 The intrinsic clearance of human recombinant CYP2C9*2 and CYP2C9*3 for the (S)-7-hydroxylation was 28.9 and 50.9% lower than of CYP2C9*1, respectively. Sulfur 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 15742978-4 2004 The intrinsic clearance of human recombinant CYP2C9*2 and CYP2C9*3 for the (S)-7-hydroxylation was 28.9 and 50.9% lower than of CYP2C9*1, respectively. Sulfur 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15742978-4 2004 The intrinsic clearance of human recombinant CYP2C9*2 and CYP2C9*3 for the (S)-7-hydroxylation was 28.9 and 50.9% lower than of CYP2C9*1, respectively. Sulfur 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15742978-6 2004 Increasing plasma AUC metabolic ratios (parent compound/metabolite) in CYP2C9*2 and CYP2C9*3 variant allele carriers were found for each hydroxylation reaction and the CYP2C9*3/*3 genotype corresponded to an about 10-fold higher metabolic ratio of PPC (S)-7-hydroxylation relative to CYP2C9*1/*1. Sulfur 252-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 15742978-6 2004 Increasing plasma AUC metabolic ratios (parent compound/metabolite) in CYP2C9*2 and CYP2C9*3 variant allele carriers were found for each hydroxylation reaction and the CYP2C9*3/*3 genotype corresponded to an about 10-fold higher metabolic ratio of PPC (S)-7-hydroxylation relative to CYP2C9*1/*1. Sulfur 252-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 15742978-6 2004 Increasing plasma AUC metabolic ratios (parent compound/metabolite) in CYP2C9*2 and CYP2C9*3 variant allele carriers were found for each hydroxylation reaction and the CYP2C9*3/*3 genotype corresponded to an about 10-fold higher metabolic ratio of PPC (S)-7-hydroxylation relative to CYP2C9*1/*1. Sulfur 252-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 15742978-6 2004 Increasing plasma AUC metabolic ratios (parent compound/metabolite) in CYP2C9*2 and CYP2C9*3 variant allele carriers were found for each hydroxylation reaction and the CYP2C9*3/*3 genotype corresponded to an about 10-fold higher metabolic ratio of PPC (S)-7-hydroxylation relative to CYP2C9*1/*1. Sulfur 252-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 15306208-4 2004 On the other hand, cisapride strongly inhibited CYP3A4 and markedly inhibited CYP2C9. Cisapride 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15247556-2 2004 Phenytoin (PHT) is a CYP3A4 inducer and CYP2C9/CYP2C19 substrate. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15181000-0 2004 The structure of human cytochrome P450 2C9 complexed with flurbiprofen at 2.0-A resolution. Flurbiprofen 58-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-42 15247556-2 2004 Phenytoin (PHT) is a CYP3A4 inducer and CYP2C9/CYP2C19 substrate. Phenytoin 11-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 15310247-8 2004 The liver microsomal SOH activity showed significant correlations with tolbutamide hydroxylation (r = 0.569) and chlorzoxazone hydroxylation (r = 0.770) activities, which were the model reactions catalyzed by CYP2C9 and CYP2E1, respectively. Tolbutamide 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 15310247-8 2004 The liver microsomal SOH activity showed significant correlations with tolbutamide hydroxylation (r = 0.569) and chlorzoxazone hydroxylation (r = 0.770) activities, which were the model reactions catalyzed by CYP2C9 and CYP2E1, respectively. Chlorzoxazone 113-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 15290664-2 2004 The cytochrome P-450 (CYP) 2C9 (CYP2C9) enzyme is involved in the metabolism of several drugs, including possibly aspirin, and such carcinogens as smoking-related polycyclic aromatic hydrocarbons. Aspirin 114-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-30 15289788-1 2004 UNLABELLED: Background and aim Previous data indicate that the urinary losartan/E-3174 ratio is a marker for cytochrome P450 (CYP) 2C9 activity in vivo. Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-134 15289788-1 2004 UNLABELLED: Background and aim Previous data indicate that the urinary losartan/E-3174 ratio is a marker for cytochrome P450 (CYP) 2C9 activity in vivo. losartan carboxylic acid 80-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-134 15289788-3 2004 METHODS: A single oral dose of losartan (25 mg) was given to 19 Beninese subjects with CYP2C9*1/*1 (n = 9), *1/*5 (n = 1), *1/*6 (n = 1), *1/*8 (n = 2), *1/*11 (n = 3), *5/*6 (n = 1), *5/*8 (n = 1), and *8/*11 (n = 1) genotypes. Losartan 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15289788-6 2004 RESULTS: The urinary losartan/E-3174 ratio in the various genotypes was as follows: 1.85 +/- 2.4 (mean +/- SD) for CYP2C9*1/*1, 14.6 for CYP2C9*1/*5, 4.2 for CYP2C9*1/*6, 188 for CYP2C9*5/*6, 11.6 for CYP2C9*5/*8, 0.44 +/- 0.13 (mean +/- SD) for CYP2C9*1/*8, 2.2 for CYP2C9*8/*11, and 5.72 +/- 4.5 (mean +/- SD) for CYP2C9*1/*11. Losartan 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 15289788-6 2004 RESULTS: The urinary losartan/E-3174 ratio in the various genotypes was as follows: 1.85 +/- 2.4 (mean +/- SD) for CYP2C9*1/*1, 14.6 for CYP2C9*1/*5, 4.2 for CYP2C9*1/*6, 188 for CYP2C9*5/*6, 11.6 for CYP2C9*5/*8, 0.44 +/- 0.13 (mean +/- SD) for CYP2C9*1/*8, 2.2 for CYP2C9*8/*11, and 5.72 +/- 4.5 (mean +/- SD) for CYP2C9*1/*11. losartan carboxylic acid 30-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 15289788-8 2004 The urinary losartan/E-3174 ratio of the single CYP2C9*1/*6 subject was higher than the 95% confidence interval of the mean of the CYP2C9*1/*1 group (0.0-3.7), whereas the metabolic ratio of the CYP2C9*8/*11 carrier was inside the 95% confidence interval of the means of the CYP2C9*1/*1 and CYP2C9*1/*11 groups (0.0-18). losartan carboxylic acid 21-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 15289789-6 2004 The main genetic factor for reduced clearance of R-(-)-ibuprofen is the CYP2C8*3 allele, whereas the clearance for S-(+)-ibuprofen is influenced by CYP2C8*3 and CYP2C9*3 alleles to a similar extent. Ibuprofen 49-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 15289789-6 2004 The main genetic factor for reduced clearance of R-(-)-ibuprofen is the CYP2C8*3 allele, whereas the clearance for S-(+)-ibuprofen is influenced by CYP2C8*3 and CYP2C9*3 alleles to a similar extent. Ibuprofen 115-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 15289789-8 2004 As compared with individuals with no mutations, individuals with the common genotype CYP2C8*1/*3 plus CYP2C9*1/*2 (19% of the population) displayed decreased ibuprofen clearance (mean, 65% [95% CI, 42%-89%]; P <.001). Ibuprofen 158-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 15289789-9 2004 Individuals homozygous or double-heterozygous for CYP2C8*3 and CYP2C9*3 variant alleles (8% of the population) had extremely low ibuprofen clearance rates, with values ranging from 7% to 27% of the mean clearance rates among noncarriers of mutations (P <.001). Ibuprofen 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15289789-11 2004 CONCLUSION: Low ibuprofen clearance occurs in a substantial proportion of healthy subjects, is not enantiospecific, and is strongly linked to CYP2C8 and CYP2C9 polymorphisms. Ibuprofen 16-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15258107-5 2004 Pantoprazole was a competitive inhibitor of both CYP2C9-catalyzed diclofenac 4"-hydroxylation and CYP3A4-catalyzed midazolam 1"-hydroxylation (K(i) of 6 and 22 microM, respectively), which were at least 2 times more potent than the other PPIs. Pantoprazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 15258107-5 2004 Pantoprazole was a competitive inhibitor of both CYP2C9-catalyzed diclofenac 4"-hydroxylation and CYP3A4-catalyzed midazolam 1"-hydroxylation (K(i) of 6 and 22 microM, respectively), which were at least 2 times more potent than the other PPIs. Diclofenac 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 15258107-7 2004 The inhibitory potency of a nonenzymatically formed product of rabeprazole, rabeprazole thioether, was also investigated and showed potent, competitive inhibition with K(i) values of 6 microM for CYP2C9, 2 to 8 microM for CYP2C19, 12 microM for CYP2D6, and 15 microM for CYP3A4. Rabeprazole 63-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 15258107-7 2004 The inhibitory potency of a nonenzymatically formed product of rabeprazole, rabeprazole thioether, was also investigated and showed potent, competitive inhibition with K(i) values of 6 microM for CYP2C9, 2 to 8 microM for CYP2C19, 12 microM for CYP2D6, and 15 microM for CYP3A4. rabeprazole-thioether 76-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 15258107-8 2004 The inhibitory potency of R-omeprazole on the four studied P450 enzymes was also studied and showed higher inhibitory potency than its S-isomer on CYP2C9 and 2C19 activities. r-omeprazole 26-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 15258107-9 2004 Our data suggest that, although the inhibitory profiles of the five studied PPIs were similar, lansoprazole and pantoprazole are the most potent in vitro inhibitors of CYP2C19 and CYP2C9, respectively. Lansoprazole 95-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 15258107-9 2004 Our data suggest that, although the inhibitory profiles of the five studied PPIs were similar, lansoprazole and pantoprazole are the most potent in vitro inhibitors of CYP2C19 and CYP2C9, respectively. Pantoprazole 112-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 15290664-2 2004 The cytochrome P-450 (CYP) 2C9 (CYP2C9) enzyme is involved in the metabolism of several drugs, including possibly aspirin, and such carcinogens as smoking-related polycyclic aromatic hydrocarbons. Aspirin 114-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15290664-2 2004 The cytochrome P-450 (CYP) 2C9 (CYP2C9) enzyme is involved in the metabolism of several drugs, including possibly aspirin, and such carcinogens as smoking-related polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 163-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-30 15290664-2 2004 The cytochrome P-450 (CYP) 2C9 (CYP2C9) enzyme is involved in the metabolism of several drugs, including possibly aspirin, and such carcinogens as smoking-related polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 163-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15122071-4 2004 Using CYP supersomes it was shown that thiocoraline is mainly metabolized by CYP3A4, with CYP1A1, CYP2C8 and CYP2C9 playing a minor role in the biotransformation (<3%). thiocoraline 39-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 15197517-0 2004 Effect of a selective CYP2C9 inhibitor on the pharmacokinetics of nateglinide in healthy subjects. Nateglinide 66-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Sulfinpyrazone 108-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Sulfinpyrazone 108-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 266-272 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Sulfinpyrazone 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Sulfinpyrazone 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 266-272 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Nateglinide 162-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Nateglinide 162-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 266-272 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Nateglinide 175-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15197517-1 2004 PURPOSE: The objective of the study was to determine the effect of a potent and selective CYP2C9 inhibitor, sulfinpyrazone (Anturane), on the pharmacokinetics of nateglinide (Starlix), a novel antidiabetic drug which is primarily (approximately 70%) metabolized via CYP2C9. Nateglinide 175-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 266-272 15285849-4 2004 Quercetin, the biflavone amentoflavone, sesamin, as well as (Z,Z)-4,4"-(1,4-pentadiene-1,5-diyl)diphenol and 3-nonadec-8-enyl-benzene-1,2-diol, were also inhibitors of CYP2C9. (z,z)-4,4"-(1,4-pentadiene-1,5-diyl)diphenol 60-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 15285849-5 2004 The IC50 of amentoflavone for CYP2C9 was 0.019 microg mL(-1) (0.035 microM). amentoflavone 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15284535-6 2004 CYP2C9*11 appeared to be a putative poor metabolizer allele, exhibiting a three-fold increase in the Km and more than a two-fold decrease in the intrinsic clearance for tolbutamide. Tolbutamide 169-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15284535-7 2004 Examination of the crystal structure of human CYP2C9 reveals that R335 is located in the turn between the J and J" helices and forms a hydrogen-bonding ion pair with D341 from the J" helix. Hydrogen 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 15284535-7 2004 Examination of the crystal structure of human CYP2C9 reveals that R335 is located in the turn between the J and J" helices and forms a hydrogen-bonding ion pair with D341 from the J" helix. d341 166-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 15284535-10 2004 A second potentially PM allele CYP2C9*12 found in a racially unidentified sample also exhibited a modest decrease in the Vmax and the intrinsic clearance for tolbutamide in a recombinant system. Tolbutamide 158-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 15284536-0 2004 Relative impact of covariates in prescribing warfarin according to CYP2C9 genotype. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 15229460-9 2004 After 7 simulated daily doses, significant differences were observed between CYP2C9*1/*1 and CYP2C9*1/*3 individuals in relation to the minimum plasma (trough) tenoxicam concentration (Cmin, 5.2 +/- 1.3 microg x mL(-1) versus 7.6 +/- 2.6 microg x mL(-1); P = .021), maximum tenoxicam plasma concentration (Cmax, 7.4 +/- 1.9 microg x mL(-1) versus 10.5 +/- 3.0 microg x mL(-1); P = .020), and 24-hour AUC (152 +/- 39 microg x mL(-1) x h versus 219 +/- 72 microg x mL(-1) x h). tenoxicam 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 15229460-0 2004 CYP2C9 genotypes and the pharmacokinetics of tenoxicam in Brazilians. tenoxicam 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15229460-1 2004 OBJECTIVE: Cytochrome P450 (CYP) 2C9, the product of the polymorphic gene CYP2C9, provides the major catabolic pathway for several anti-inflammatory drugs, including tenoxicam. tenoxicam 166-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-36 15229460-1 2004 OBJECTIVE: Cytochrome P450 (CYP) 2C9, the product of the polymorphic gene CYP2C9, provides the major catabolic pathway for several anti-inflammatory drugs, including tenoxicam. tenoxicam 166-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15229460-8 2004 After a single dose of tenoxicam, the area under the plasma concentration-time curve (AUC) from time 0 to infinity and the oral clearance of tenoxicam were 190 +/- 48 microg x mL(-1) x h and 113 +/- 30 mL x h(-1), respectively, in wild-type homozygous subjects (CYP2C9*1/*1), as compared with 261 +/- 14 microg x mL(-1) x h (P = .013) and 77 +/- 4 mL x h(-1) (P = .036), respectively, in CYP2C9*1/*2 heterozygous subjects and 335 +/- 126 microg x mL(-1) x h (P = .003) and 67 +/- 23 mL x h(-1) (P = .008) in CYP2C9*1/*3, respectively, heterozygous subjects. tenoxicam 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 262-268 15229460-8 2004 After a single dose of tenoxicam, the area under the plasma concentration-time curve (AUC) from time 0 to infinity and the oral clearance of tenoxicam were 190 +/- 48 microg x mL(-1) x h and 113 +/- 30 mL x h(-1), respectively, in wild-type homozygous subjects (CYP2C9*1/*1), as compared with 261 +/- 14 microg x mL(-1) x h (P = .013) and 77 +/- 4 mL x h(-1) (P = .036), respectively, in CYP2C9*1/*2 heterozygous subjects and 335 +/- 126 microg x mL(-1) x h (P = .003) and 67 +/- 23 mL x h(-1) (P = .008) in CYP2C9*1/*3, respectively, heterozygous subjects. tenoxicam 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 388-394 15229460-8 2004 After a single dose of tenoxicam, the area under the plasma concentration-time curve (AUC) from time 0 to infinity and the oral clearance of tenoxicam were 190 +/- 48 microg x mL(-1) x h and 113 +/- 30 mL x h(-1), respectively, in wild-type homozygous subjects (CYP2C9*1/*1), as compared with 261 +/- 14 microg x mL(-1) x h (P = .013) and 77 +/- 4 mL x h(-1) (P = .036), respectively, in CYP2C9*1/*2 heterozygous subjects and 335 +/- 126 microg x mL(-1) x h (P = .003) and 67 +/- 23 mL x h(-1) (P = .008) in CYP2C9*1/*3, respectively, heterozygous subjects. tenoxicam 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 388-394 15205386-3 2004 In the present study, in vitro enzyme kinetic data were used to predict the in vivo clearance and drug-drug interaction potential of four well known CYP2C9 substrates (tolbutamide, fluvastatin, ibuprofen and diclofenac) that are frequently used as benchmark substances in screening programs. Tolbutamide 168-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15205386-3 2004 In the present study, in vitro enzyme kinetic data were used to predict the in vivo clearance and drug-drug interaction potential of four well known CYP2C9 substrates (tolbutamide, fluvastatin, ibuprofen and diclofenac) that are frequently used as benchmark substances in screening programs. Fluvastatin 181-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15205386-3 2004 In the present study, in vitro enzyme kinetic data were used to predict the in vivo clearance and drug-drug interaction potential of four well known CYP2C9 substrates (tolbutamide, fluvastatin, ibuprofen and diclofenac) that are frequently used as benchmark substances in screening programs. Ibuprofen 194-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15205386-3 2004 In the present study, in vitro enzyme kinetic data were used to predict the in vivo clearance and drug-drug interaction potential of four well known CYP2C9 substrates (tolbutamide, fluvastatin, ibuprofen and diclofenac) that are frequently used as benchmark substances in screening programs. Diclofenac 208-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15197523-2 2004 Losartan has recently been suggested as a selective probe for CYP2C9 metabolic activity. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15197523-3 2004 The aim of the study was to determine the activity of CYP2C9, using losartan as a probe drug, in relation to CYP2C9 genotype in healthy Turkish subjects. Losartan 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 15197523-3 2004 The aim of the study was to determine the activity of CYP2C9, using losartan as a probe drug, in relation to CYP2C9 genotype in healthy Turkish subjects. Losartan 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 15197523-6 2004 Losartan oxidation was also studied in vitro, using human CYP2C8 and CYP2C9 enzymes expressed in yeast. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 15197523-8 2004 The urinary losartan/E3174 ratio was significantly higher in subjects with CYP2C9*1/*3 genotype (median 2.35, n=12) than in subjects with CYP2C9*1/*1 (0.71, n=58) and *1/*2 (0.85, n=10) genotypes ( P<0.05). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 15197523-8 2004 The urinary losartan/E3174 ratio was significantly higher in subjects with CYP2C9*1/*3 genotype (median 2.35, n=12) than in subjects with CYP2C9*1/*1 (0.71, n=58) and *1/*2 (0.85, n=10) genotypes ( P<0.05). Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15197523-8 2004 The urinary losartan/E3174 ratio was significantly higher in subjects with CYP2C9*1/*3 genotype (median 2.35, n=12) than in subjects with CYP2C9*1/*1 (0.71, n=58) and *1/*2 (0.85, n=10) genotypes ( P<0.05). losartan carboxylic acid 21-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 15197523-10 2004 CONCLUSION: The urinary losartan to E3174 metabolic ratio after a 25-mg losartan dose was found to be a safe and useful phenotyping assay for CYP2C9 activity in vivo. Losartan 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 15197523-11 2004 CYP2C9*3 variant allele is a major determinant of the enzyme activity, and it decreases losartan metabolism significantly, while CYP2C9*2 allele has less impact on enzyme function. Losartan 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15197523-11 2004 CYP2C9*3 variant allele is a major determinant of the enzyme activity, and it decreases losartan metabolism significantly, while CYP2C9*2 allele has less impact on enzyme function. Losartan 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 15349140-2 2004 CYP2C9*2 (Arg144Cys) and *3 (Ile359Leu) have been the most widely studied alleles. ile359leu 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15349140-5 2004 In addition, multiple human studies have demonstrated significant associations between CYP2C9 genotype and the disposition of substrates such as warfarin, phenytoin and various sulfonylureas, angiotensin II receptor blockers and non-steroidal antiinflammatory agents. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15349140-5 2004 In addition, multiple human studies have demonstrated significant associations between CYP2C9 genotype and the disposition of substrates such as warfarin, phenytoin and various sulfonylureas, angiotensin II receptor blockers and non-steroidal antiinflammatory agents. Phenytoin 155-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15349140-5 2004 In addition, multiple human studies have demonstrated significant associations between CYP2C9 genotype and the disposition of substrates such as warfarin, phenytoin and various sulfonylureas, angiotensin II receptor blockers and non-steroidal antiinflammatory agents. Sulfonylurea Compounds 177-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 15349140-6 2004 Individuals carrying the CYP2C9*2 and *3 alleles also have lower warfarin and phenytoin daily dose requirements, and appear more susceptible to adverse events during the initiation of therapy. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 15349140-6 2004 Individuals carrying the CYP2C9*2 and *3 alleles also have lower warfarin and phenytoin daily dose requirements, and appear more susceptible to adverse events during the initiation of therapy. Phenytoin 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 15226678-1 2004 OBJECTIVES: Cytochrome P450 (CYP) 2C9 metabolizes about 16% of drugs in current clinical use, including lornoxicam and tolbutamide. lornoxicam 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 15226678-1 2004 OBJECTIVES: Cytochrome P450 (CYP) 2C9 metabolizes about 16% of drugs in current clinical use, including lornoxicam and tolbutamide. Tolbutamide 119-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 15213846-0 2004 The risk of bleeding complications in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Acenocoumarol 100-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 15213846-0 2004 The risk of bleeding complications in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Acenocoumarol 100-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15213846-0 2004 The risk of bleeding complications in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Phenprocoumon 117-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 15213846-1 2004 The principal enzyme involved in coumarin metabolism is CYP2C9. coumarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 15213846-4 2004 It was, therefore, our objective to study the effect of the CYP2C9 polymorphisms on bleeding complications during initiation and maintenance phases of coumarin anticoagulant therapy. coumarin 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 15213846-15 2004 In this study among outpatients of an anticoagulation clinic using acenocoumarol or phenprocoumon, having a variant allele of CYP2C9 was associated with an increased risk of major bleeding events in patients on acenocoumarol, but not in patients on phenprocoumon. Acenocoumarol 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 15213846-15 2004 In this study among outpatients of an anticoagulation clinic using acenocoumarol or phenprocoumon, having a variant allele of CYP2C9 was associated with an increased risk of major bleeding events in patients on acenocoumarol, but not in patients on phenprocoumon. Phenprocoumon 84-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 15213846-15 2004 In this study among outpatients of an anticoagulation clinic using acenocoumarol or phenprocoumon, having a variant allele of CYP2C9 was associated with an increased risk of major bleeding events in patients on acenocoumarol, but not in patients on phenprocoumon. Acenocoumarol 211-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 15213846-15 2004 In this study among outpatients of an anticoagulation clinic using acenocoumarol or phenprocoumon, having a variant allele of CYP2C9 was associated with an increased risk of major bleeding events in patients on acenocoumarol, but not in patients on phenprocoumon. Phenprocoumon 249-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 15213846-16 2004 Although one might consider the assessment of the CYP2C9 genotype of a patient for dose adjustment before starting treatment with acenocoumarol, a prospective randomised trial should demonstrate whether this reduces the increased risk of major bleeding events. Acenocoumarol 130-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 15229460-9 2004 After 7 simulated daily doses, significant differences were observed between CYP2C9*1/*1 and CYP2C9*1/*3 individuals in relation to the minimum plasma (trough) tenoxicam concentration (Cmin, 5.2 +/- 1.3 microg x mL(-1) versus 7.6 +/- 2.6 microg x mL(-1); P = .021), maximum tenoxicam plasma concentration (Cmax, 7.4 +/- 1.9 microg x mL(-1) versus 10.5 +/- 3.0 microg x mL(-1); P = .020), and 24-hour AUC (152 +/- 39 microg x mL(-1) x h versus 219 +/- 72 microg x mL(-1) x h). tenoxicam 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15229460-12 2004 Heterozygosis for CYP2C9*3, and to a lesser degree CYP2C9*2, increases the exposure to tenoxicam during single and repeated doses. tenoxicam 87-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15229460-12 2004 Heterozygosis for CYP2C9*3, and to a lesser degree CYP2C9*2, increases the exposure to tenoxicam during single and repeated doses. tenoxicam 87-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 15180495-4 2004 With in vivo animal models, acute moderate hypoxia (PaO2 of around 35-50 mm Hg) reduces the clearance of drugs biotransformed by CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2E1, although hypoxia does not affect the clearance of drugs biotransformed by CYP3A6. pao2 52-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15672756-6 2004 The relative activity factor approach assumed that NE-098 formation is predominantly catalysed by CYP3A4 and CYP2C9 and the NE-152+163mix (a mixture of two hydroxylated metabolites, NE-152 and NE-163) formation is only catalysed by CYP3A4. CHEMBL542786 51-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 15196026-5 2004 Torsemide is preferentially metabolized by CYP2C9, and it was anticipated that the mutations in CYP2C8 might increase activity. Torsemide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 15196026-9 2004 Mutation of CYP2C8 at position 359 (S359I), a site of genetic polymorphism in CYP2C9, resulted in relatively minor changes in paclitaxel- and torsemide-hydroxylase activities. Paclitaxel 126-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15170332-0 2004 Quantitative binding models for CYP2C9 based on benzbromarone analogues. Benzbromarone 48-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15170332-2 2004 To uncover the determinants of specificity for cytochrome CYP2C9, a novel library of benzbromarone (bzbr) inhibitors was used to reevaluate its pharmacophore. Benzbromarone 85-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15170332-2 2004 To uncover the determinants of specificity for cytochrome CYP2C9, a novel library of benzbromarone (bzbr) inhibitors was used to reevaluate its pharmacophore. Benzbromarone 100-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Flurbiprofen 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Flurbiprofen 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Dapsone 100-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Dapsone 100-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Flurbiprofen 147-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15170358-1 2004 Cytochrome P450 2C9 (CYP2C9)-mediated flurbiprofen 4"-hydroxylation is activated by the presence of dapsone resulting in reduction of the K(m) for flurbiprofen hydroxylation and an increase in V(m). Flurbiprofen 147-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15170358-2 2004 Previous spectral binding studies have demonstrated that the binding of flurbiprofen with CYP2C9 is increased (decrease in K(S)) by the presence of dapsone. Flurbiprofen 72-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15170358-2 2004 Previous spectral binding studies have demonstrated that the binding of flurbiprofen with CYP2C9 is increased (decrease in K(S)) by the presence of dapsone. Dapsone 148-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 15170358-9 2004 Molecular modeling studies were also performed to corroborate the relative orientations of flurbiprofen and dapsone in the active site of CYP2C9. Flurbiprofen 91-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15170358-9 2004 Molecular modeling studies were also performed to corroborate the relative orientations of flurbiprofen and dapsone in the active site of CYP2C9. Dapsone 108-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15170358-10 2004 Shift of the 4" proton of flurbiprofen closer to the heme iron of CYP2C9 in the presence of dapsone may play a role in activation. Flurbiprofen 26-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 15170358-10 2004 Shift of the 4" proton of flurbiprofen closer to the heme iron of CYP2C9 in the presence of dapsone may play a role in activation. Heme 53-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 15170358-10 2004 Shift of the 4" proton of flurbiprofen closer to the heme iron of CYP2C9 in the presence of dapsone may play a role in activation. Iron 58-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 15170358-10 2004 Shift of the 4" proton of flurbiprofen closer to the heme iron of CYP2C9 in the presence of dapsone may play a role in activation. Dapsone 92-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 15145963-2 2004 In addition, warfarin S/R ratios are used to assess CYP2C9 activity. Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 15145963-11 2004 Single-point and two-point S-warfarin concentrations, but not warfarin S/R ratios, were predictive of S-warfarin AUC(0- infinity ) during CYP2C9 baseline activity and inhibition. Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15145963-14 2004 LSS using S-warfarin concentrations is an efficient and accurate technique to evaluate S-warfarin AUC(0- infinity ) when using warfarin as a CYP2C9 probe drug. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15145963-14 2004 LSS using S-warfarin concentrations is an efficient and accurate technique to evaluate S-warfarin AUC(0- infinity ) when using warfarin as a CYP2C9 probe drug. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 15046773-6 2004 Following exposure to OTA, cells expressing CYP2C9 showed a significant reduction in neutral red (NR) uptake but not in Alamar blue (AB) reduction, as compared to the control LNCX cells which do not express CYP450 enzymes. Neutral Red 85-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. L-Buthionine 22-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. L-Buthionine 22-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. r-sulphoximine 37-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. r-sulphoximine 37-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. Buthionine Sulfoximine 53-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. Buthionine Sulfoximine 53-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 15046773-8 2004 When pre-treated with l-buthionine S,R-sulphoximine (BSO) to deplete GSH, CYP2C9-expressing cells showed also a loss of cell viability as compared to LNCX cells, although to a lesser extent as compared to non-depleted CYP2C9-expressing cells. Glutathione 69-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15175798-1 2004 Two cytochrome P450 2C9 (CYP2C9) polymorphisms, CYP2C9*2 and *3, metabolize warfarin inefficiently. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-23 15175798-1 2004 Two cytochrome P450 2C9 (CYP2C9) polymorphisms, CYP2C9*2 and *3, metabolize warfarin inefficiently. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 15175798-1 2004 Two cytochrome P450 2C9 (CYP2C9) polymorphisms, CYP2C9*2 and *3, metabolize warfarin inefficiently. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 15175798-6 2004 The CYP2C9 polymorphisms independently predicted low warfarin requirements after adjusting for Body Mass Index, age, acetaminophen use, and race (OR 24.80; 95% CI 3.83-160.78). Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 15175798-6 2004 The CYP2C9 polymorphisms independently predicted low warfarin requirements after adjusting for Body Mass Index, age, acetaminophen use, and race (OR 24.80; 95% CI 3.83-160.78). Acetaminophen 117-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 15175798-10 2004 In conclusion, CYP2C9*2 or *3 compound heterozygotes and homozygotes have low warfarin requirements even after excluding liver disease, excessive alcohol or acetaminophen consumption, low body weight, advancing age, and drug interactions. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 15175798-10 2004 In conclusion, CYP2C9*2 or *3 compound heterozygotes and homozygotes have low warfarin requirements even after excluding liver disease, excessive alcohol or acetaminophen consumption, low body weight, advancing age, and drug interactions. Alcohols 146-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 15175798-10 2004 In conclusion, CYP2C9*2 or *3 compound heterozygotes and homozygotes have low warfarin requirements even after excluding liver disease, excessive alcohol or acetaminophen consumption, low body weight, advancing age, and drug interactions. Acetaminophen 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 15056479-7 2004 CYP2C9, CYP2E1 CYP2C19 and CYP2D6 are engaged to a lesser degree in 5-sulphoxidation, while CYP1A2, CYP3A4 and CYP2D6 in perazine N-demethylation (6-10%, depending on the isoform). Perazine 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15130760-0 2004 Differential activation of CYP2C9 variants by dapsone. Dapsone 46-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 15130760-1 2004 Studies have shown that CYP2C9.1 mediated metabolism of flurbiprofen or naproxen is activated by co-incubation with dapsone. Flurbiprofen 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 15130760-1 2004 Studies have shown that CYP2C9.1 mediated metabolism of flurbiprofen or naproxen is activated by co-incubation with dapsone. Naproxen 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 15130760-1 2004 Studies have shown that CYP2C9.1 mediated metabolism of flurbiprofen or naproxen is activated by co-incubation with dapsone. Dapsone 116-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Flurbiprofen 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Flurbiprofen 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Flurbiprofen 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-4 2004 Dapsone increased the efficiency (V(m)/K(m)) of flurbiprofen 4"-hydroxylation by CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5 by 8-, 31-, 47-, and 22-fold, respectively. Flurbiprofen 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Naproxen 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Dapsone 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Dapsone 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Dapsone 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Dapsone 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Naproxen 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Naproxen 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Naproxen 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-5 2004 In similar experiments using the substrate naproxen, dapsone increased the efficiency of naproxen demethylation 7-, 15-, 13-, and 22-fold, in CYP2C9.1, CYP2C9.2, CYP2C9.3, and CYP2C9.5, respectively. Naproxen 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Dapsone 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Dapsone 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Dapsone 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Dapsone 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Naproxen 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Naproxen 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Naproxen 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-6 2004 Also, dapsone normalized naproxen"s kinetic profile from biphasic (CYP2C9.1 and CYP2C9.2) or linear (CYP2C9.3 and CYP2C9.5) to hyperbolic for all variant forms. Naproxen 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 15130760-7 2004 Thus, amino acid substitutions of CYP2C9 variants affect the degree of dapsone activation in a genotype-dependent fashion. Dapsone 71-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 15133536-3 2004 Tolbutamide 4-methylhydroxylation and testosterone 6beta-hydroxylation were used as index reactions of CYP2C9 or CYP3A4 catalytic activities, respectively. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 15133536-8 2004 Ginsenoside Rd also had significant inhibitory potency on both CYP2C9- and CYP3A4-mediated index reactions with IC(50) values of 105 and 62 micromol/L, respectively. Ginsenosides 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15133536-11 2004 Our findings suggest that quercetin and ginsenoside Rd have the potential to interact with conventional medicines that are metabolized by CYP2C9 and CYP3A4 in vivo. Quercetin 26-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15133536-11 2004 Our findings suggest that quercetin and ginsenoside Rd have the potential to interact with conventional medicines that are metabolized by CYP2C9 and CYP3A4 in vivo. Ginsenosides 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 15039471-0 2004 Warfarin resistance with poor CYP2C9 activity and CYP2C9*1*2 genotype. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15039471-0 2004 Warfarin resistance with poor CYP2C9 activity and CYP2C9*1*2 genotype. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 15108219-8 2004 Thus, DEX interacts with CYP2C9 both as inhibitor (K(i)=69.0 microM) and as substrate in vitro. dexloxiglumide 6-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 15116049-0 2004 CYP2C9 and oral anticoagulation therapy with acenocoumarol and warfarin: similarities yet differences. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15116052-1 2004 OBJECTIVE: Our objective was to assess whether there is an association between the presence of allelic variants of the gene for cytochrome P450 (CYP) 2C9 and anticoagulation problems during the initial 3 to 6 months of acenocoumarol treatment. Acenocoumarol 219-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-153 15116052-10 2004 CYP2C9 genotyping could be useful to identify potential candidates for more frequent INR controls to minimize problems with acenocoumarol anticoagulation status. Acenocoumarol 124-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15116053-1 2004 OBJECTIVE: The aim of this study was to investigate the respective contribution of the different cytochrome P450 (CYP) 2C9 genetic polymorphisms to the interindividual variability of acenocoumarol pharmacodynamic response. Acenocoumarol 183-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-122 15116053-4 2004 Finally, CYP2C9 phenotype was evaluated after a single oral dose of 4 mg of acenocoumarol. Acenocoumarol 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 15116053-6 2004 RESULTS: The CYP2C9*3 allele was the only nonsynonymous single nucleotide polymorphism (SNP) influencing acenocoumarol pharmacodynamics; the percentages of remaining factor VII were 60% +/- 19%, 39% +/- 17%, and 17% for CYP2C9*1/CYP2C9*1, CYP2C9*1/CYP2C9*3, and CYP2C9*3/CYP2C9*3 subjects, respectively (P =.001). Acenocoumarol 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15116053-9 2004 The haplotype that contains the CYP2C9*3 allele was the only one influencing acenocoumarol pharmacodynamics, explaining 14.3% of its interindividual variability. Acenocoumarol 77-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15116053-11 2004 CONCLUSION: Overall, CYP2C9-related genetic variability accounts for 14% of the interindividual variability in acenocoumarol pharmacodynamic response. Acenocoumarol 111-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 15100169-5 2004 In CYP2C9 phenotyping with losartan, three subpopulations were distinguished that differed in the number of CYP2C9*3 alleles (0, 1, or 2). Losartan 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 15100169-6 2004 A three-fold higher metabolic ratio (MR; urinary losartan/carboxymetabolite) was found comparing CYP2C9*1/*3 (n = 20) to CYP2C9*1/*1 (n = 81), but there was considerable variation within each genotype. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 15100169-6 2004 A three-fold higher metabolic ratio (MR; urinary losartan/carboxymetabolite) was found comparing CYP2C9*1/*3 (n = 20) to CYP2C9*1/*1 (n = 81), but there was considerable variation within each genotype. Losartan 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 15100169-7 2004 Subjects genotyped as CYP2C9*1/*1, but with an unexpectedly slow oxidation of losartan, were selected for DNA-sequencing analysis of the CYP2C9 gene. Losartan 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 15100169-9 2004 However, sequencing of the CYP2C9 gene was also carried out in patients genotyped as CYP2C9*1/*1 but with an exceptionally low steady-state clearance of S-warfarin. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 15100169-9 2004 However, sequencing of the CYP2C9 gene was also carried out in patients genotyped as CYP2C9*1/*1 but with an exceptionally low steady-state clearance of S-warfarin. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 15370959-6 2004 Both pranlukast and zafirlukast showed moderate inhibition of CYP2C9-catalysed tolbutamide 4-methylhydroxylation, competitively inhibiting tolbutamide 4-methylhydroxylation with estimated mean K(i) values of 3.82 +/- 0.50 and 5.86 +/- 0.08 microM, respectively. pranlukast 5-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15024534-0 2004 Genetic polymorphism of cytochrome P450 2C9 in diphenylhydantoin-induced cutaneous adverse drug reactions. Phenytoin 47-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-43 15024534-3 2004 Genetic polymorphisms leading to altered metabolic processes of cytochrome P(450) (CYP) 2C9, a main metabolic enzyme for DPH, may be the pathological mechanism for certain cases of DPH-induced CADRs. Phenytoin 121-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-91 15024534-3 2004 Genetic polymorphisms leading to altered metabolic processes of cytochrome P(450) (CYP) 2C9, a main metabolic enzyme for DPH, may be the pathological mechanism for certain cases of DPH-induced CADRs. Phenytoin 181-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-91 15024534-4 2004 OBJECTIVE: To examine the effects of an altered CYP2C9 variant, CYP2C9*3, on DPH-induced CADRs. Phenytoin 77-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 15024534-4 2004 OBJECTIVE: To examine the effects of an altered CYP2C9 variant, CYP2C9*3, on DPH-induced CADRs. Phenytoin 77-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 15024534-5 2004 METHODS: Ten patients with DPH-induced CADRs were examined for CYP2C9 genetic polymorphisms. Phenytoin 27-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15024534-8 2004 RESULTS: A heterozygous CYP2C9*3 variant was found in three of the 10 DPH-induced CADR patients. Phenytoin 70-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 15024534-13 2004 CONCLUSION: A CYP2C9*3 variant could play a role in the proportion of patients with DPH-induced CADRs that differ from patients with DPH-induced CADRs showing positive patch-test results. Phenytoin 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 15024534-13 2004 CONCLUSION: A CYP2C9*3 variant could play a role in the proportion of patients with DPH-induced CADRs that differ from patients with DPH-induced CADRs showing positive patch-test results. Phenytoin 133-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 15045499-0 2004 Cytochrome P450 2C9 phenotyping using low-dose tolbutamide. Tolbutamide 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 15045499-1 2004 OBJECTIVES: The hypoglycaemic drug tolbutamide is used for assessment of CYP2C9 activity in vivo. Tolbutamide 35-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 15045499-2 2004 However, therapeutically active doses of 500 mg bear the risk of hypoglycaemia, and a tolbutamide-derived parameter based on a single plasma or urine concentration reflecting CYP2C9 activity accurately is lacking. Tolbutamide 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 15045499-9 2004 CONCLUSIONS: A low dose of 125 mg tolbutamide can safely and accurately be used for CYP2C9 phenotyping. Tolbutamide 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 15045499-10 2004 As a simple metric for CYP2C9 activity, we propose to determine tolbutamide in plasma 24 h after drug intake. Tolbutamide 64-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 15118259-6 2004 (14)C-Pitavastatin is metabolized with CYP2C9 to 8-hydroxy derivative, but its Vmax /Km was about 2 micro l/min/mg, about 1/8 to 1/100 in comparison to the reported values of other statins, indicating that pitavastatin is hardly metabolized. c-pitavastatin 4-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 15370959-6 2004 Both pranlukast and zafirlukast showed moderate inhibition of CYP2C9-catalysed tolbutamide 4-methylhydroxylation, competitively inhibiting tolbutamide 4-methylhydroxylation with estimated mean K(i) values of 3.82 +/- 0.50 and 5.86 +/- 0.08 microM, respectively. zafirlukast 20-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15370959-6 2004 Both pranlukast and zafirlukast showed moderate inhibition of CYP2C9-catalysed tolbutamide 4-methylhydroxylation, competitively inhibiting tolbutamide 4-methylhydroxylation with estimated mean K(i) values of 3.82 +/- 0.50 and 5.86 +/- 0.08 microM, respectively. Tolbutamide 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15370959-6 2004 Both pranlukast and zafirlukast showed moderate inhibition of CYP2C9-catalysed tolbutamide 4-methylhydroxylation, competitively inhibiting tolbutamide 4-methylhydroxylation with estimated mean K(i) values of 3.82 +/- 0.50 and 5.86 +/- 0.08 microM, respectively. Tolbutamide 139-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 15370959-12 2004 The results suggest that like zafirlukast, pranlukast also has the potential moderately to inhibit CYP2C9-catalysed tolbutamide 4-methylhydroxylation. pranlukast 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 15370959-12 2004 The results suggest that like zafirlukast, pranlukast also has the potential moderately to inhibit CYP2C9-catalysed tolbutamide 4-methylhydroxylation. Tolbutamide 116-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 15370959-13 2004 Therefore, the inhibitory potential of pranlukast should be considered when it is co-administered with CYP2C9 substrates with narrow therapeutic ranges (e.g. S-warfarin, phenytoin). pranlukast 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 15370961-0 2004 Stereoselective glucuronidation and hydroxylation of etodolac by UGT1A9 and CYP2C9 in man. Etodolac 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 15370961-8 2004 Of several human P450 enzymes, CYP2C9 had the greatest activity for hydroxylation of R-etodolac. Etodolac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 15370961-9 2004 Sulfaphenazole, an inhibitor of CYP2C9, and anti-CYP2C9 antibody inhibited the hydroxylation of R-etodolac in human liver microsomes. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15370961-9 2004 Sulfaphenazole, an inhibitor of CYP2C9, and anti-CYP2C9 antibody inhibited the hydroxylation of R-etodolac in human liver microsomes. Etodolac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15370961-9 2004 Sulfaphenazole, an inhibitor of CYP2C9, and anti-CYP2C9 antibody inhibited the hydroxylation of R-etodolac in human liver microsomes. Etodolac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 15370961-10 2004 CYP2C9 therefore contributes to the stereoselective hydroxylation of R-etodolac. Etodolac 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 14656880-0 2004 Association of pharmacokinetic (CYP2C9) and pharmacodynamic (factors II, VII, IX, and X; proteins S and C; and gamma-glutamyl carboxylase) gene variants with warfarin sensitivity. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15070684-0 2004 5"-Flanking region polymorphisms of CYP2C9 and their relationship to S-warfarin metabolism in white and Japanese patients. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 15070684-1 2004 White and Japanese patients require different warfarin dosages to achieve therapeutic anticoagulation, but this can be only partly explained by genetic variability in the coding region of CYP2C9-a critical enzyme in the drug"s metabolism. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 15070684-2 2004 Accordingly, analysis of the -2.1-kb 5"-flanking region of CYP2C9 was undertaken in 22 white and 38 Japanese patients whose unbound oral clearance of S-warfarin had been previously determined. Warfarin 150-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 15135088-11 2004 Diclofenac efficiently inhibited CYP2C9 and to a less extent CYP3A4 enzyme. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15094935-1 2004 The objective of this study was to report 2 cases of CYP2C9 genetic polymorphism and elevated warfarin S:R ratios in patients taking low doses of warfarin, and compare the observed characteristics with those in published reports. Warfarin 146-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 15094935-6 2004 This report of a CYP2C9*3 heterozygous individual taking a low dose of warfarin is consistent with previous reports in the literature. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 15094935-7 2004 This summary of a CYP2C9*6 homozygous individual taking a low dose of warfarin is the first such published report. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 15094935-8 2004 CYP2C9 genotyping in these patients provided a likely explanation for their continued low warfarin dosage requirements. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15094935-9 2004 Awareness of a patient"s CYP2C9 genotype may provide an explanation for low warfarin dosage requirements in stable patients and may help in determining the optimal dose in patients being initiated on warfarin. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 15094935-9 2004 Awareness of a patient"s CYP2C9 genotype may provide an explanation for low warfarin dosage requirements in stable patients and may help in determining the optimal dose in patients being initiated on warfarin. Warfarin 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 14656880-6 2004 Multiple regression analysis revealed that warfarin sensitivity was independently associated with -402G-->A, (CAA repeat)n, CYP2C9*3, and 165Thr-->Met, which accounted for 50% of variance. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 15051744-12 2004 Since this isozyme had the lowest in vitro IC(50) values for the cytochrome P450s most commonly involved with the metabolism of drugs (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4), azimilide-related drug interactions mediated via these isozymes are not anticipated. azimilide 181-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 15039296-0 2004 Assessment of arginine 97 and lysine 72 as determinants of substrate specificity in cytochrome P450 2C9 (CYP2C9). Arginine 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 15039296-0 2004 Assessment of arginine 97 and lysine 72 as determinants of substrate specificity in cytochrome P450 2C9 (CYP2C9). Arginine 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 15039296-0 2004 Assessment of arginine 97 and lysine 72 as determinants of substrate specificity in cytochrome P450 2C9 (CYP2C9). Lysine 30-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 15039296-0 2004 Assessment of arginine 97 and lysine 72 as determinants of substrate specificity in cytochrome P450 2C9 (CYP2C9). Lysine 30-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 14739663-4 2004 The major enzyme involved in the metabolism of acenocoumarol in man is CYP2C9. Acenocoumarol 47-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 15050794-1 2004 PURPOSE: Allelic variants of the gene coding for cytochrome P450 isoform 2C9 (CYP2C9), 2C9*2 and 2C9*3, were shown to increase sensitivity to warfarin in adults. Warfarin 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-76 14722322-9 2004 Treatment with 10 microM rifampin resulted in increases in CYP3A4 mRNA (17-fold) and activity (6-beta-hydroxytestoterone formation, 9-fold); and in CYP2C9 mRNA (4-fold) and activity (4"-hydroxydiclofenac formation, 2-fold). Rifampin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 15050794-1 2004 PURPOSE: Allelic variants of the gene coding for cytochrome P450 isoform 2C9 (CYP2C9), 2C9*2 and 2C9*3, were shown to increase sensitivity to warfarin in adults. Warfarin 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 15050794-3 2004 The aim of our purpose was to investigate whether a genetic factor, such as cyp2c9 genotype, does influence the warfarin maintenance dose in very elderly patients. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 15001966-12 2004 CONCLUSION: This study showed that rifampin affected the disposition of rosiglitazone in humans, probably by the induction of cytochrome P450 (CYP) 2C8 and, to a lesser extent, CYP2C9. Rifampin 35-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 15268978-10 2004 Recombinant CYP3A4, CYP3A5, CYP2C9 and CYP2D6, but not CYP1A2, catalysed norhydromorphone formation, whereas none of these enzymes was active in dihydroisomorphine formation. norhydromorphone 73-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 15268978-10 2004 Recombinant CYP3A4, CYP3A5, CYP2C9 and CYP2D6, but not CYP1A2, catalysed norhydromorphone formation, whereas none of these enzymes was active in dihydroisomorphine formation. Dihydroisomorphine 145-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 15268978-12 2004 In summary, CYP3A and, to a lesser extent, CYP2C9 catalysed hydromorphone N-demethylation in human liver microsomes. Hydromorphone 60-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 15001966-12 2004 CONCLUSION: This study showed that rifampin affected the disposition of rosiglitazone in humans, probably by the induction of cytochrome P450 (CYP) 2C8 and, to a lesser extent, CYP2C9. Rosiglitazone 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 15001971-1 2004 OBJECTIVE: Variant alleles of the CYP2C9 gene encoding the cytochrome P450 (CYP) enzyme (2C9*2 [Arg144Cys] and 2C9*3 [Ile359Leu]) are known to increase the anticoagulant effect of warfarin and decrease the mean daily dose required to maintain the international normalized ratio (INR) of the prothrombin time within the target therapeutic range. Warfarin 180-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 15001971-7 2004 CONCLUSIONS: The requirement of smaller doses of warfarin in relation to CYP2C9 polymorphisms is already manifest on the fourth day of treatment, at the time of the first INR estimate. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 15001972-0 2004 Contribution of age, body size, and CYP2C9 genotype to anticoagulant response to warfarin. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 15001972-1 2004 OBJECTIVE: Our objective was to assess the contribution of CYP2C9 genotype, age, body size, and vitamin K and lipid status to warfarin dose requirements. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 15001972-10 2004 CYP2C9 genotype had a significant effect on S-warfarin clearance (r = 0.34, P <.0001) but none on R-warfarin clearance. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15001972-11 2004 CONCLUSION: This study showed that age and CYP2C9 polymorphism affect warfarin dose requirements in patients receiving long-term therapy and having stable control of anticoagulation. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 15001972-12 2004 It is anticipated that using dosing regimens modified to take into account the contribution of age and CYP2C9 genotype has the potential to improve the safety of warfarin therapy. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 15058617-6 2004 Other dental therapeutic agents are substrates for CYP2C9 (celecoxib, ibuprofen and naproxen), CYP2D6 (codeine and tramadol), CYP3A4 (methylprednisolone) and CYP2E1 (acetaminophen). Celecoxib 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 14977868-7 2004 Moreover, sulfaphenazole, a CYP2C9 competitive inhibitor, inhibited disulfoton sulfoxidation by CYP2C9 (IC50 0.84 microM) as well as in HLMs. Sulfaphenazole 10-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 14977868-7 2004 Moreover, sulfaphenazole, a CYP2C9 competitive inhibitor, inhibited disulfoton sulfoxidation by CYP2C9 (IC50 0.84 microM) as well as in HLMs. Sulfaphenazole 10-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 14977868-7 2004 Moreover, sulfaphenazole, a CYP2C9 competitive inhibitor, inhibited disulfoton sulfoxidation by CYP2C9 (IC50 0.84 microM) as well as in HLMs. Disulfoton 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 14977868-7 2004 Moreover, sulfaphenazole, a CYP2C9 competitive inhibitor, inhibited disulfoton sulfoxidation by CYP2C9 (IC50 0.84 microM) as well as in HLMs. Disulfoton 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 15048614-3 2004 The present study was aimed to evaluate the possible inhibition of CYP2D6 during haloperidol treatment, and to determine the effect of CYP2D6 and CYP2C9 genotypes on the plasma concentration of haloperidol. Haloperidol 194-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 14718257-0 2004 5-Hydroxytryptamine is biotransformed by CYP2C9, 2C19 and 2B6 to hydroxylamine, which is converted into nitric oxide. Serotonin 0-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 14970873-8 2004 In conclusion, the detected presence of CYP3A4 and CYP2C9 in breast tumours offers the possibility of intratumoral turnover of ifosfamide. Ifosfamide 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 14717783-6 2004 There was no correlation between warfarin daily dose and average daily dietary vitamin K intake when calculated over 28 d. The regression model for warfarin dose showed that, while dietary vitamin K had no effect, CYP2C9 genotype (P = 2%) and age (P < 1%) significantly contributed to inter-patient variability in warfarin dose requirements. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 14717783-6 2004 There was no correlation between warfarin daily dose and average daily dietary vitamin K intake when calculated over 28 d. The regression model for warfarin dose showed that, while dietary vitamin K had no effect, CYP2C9 genotype (P = 2%) and age (P < 1%) significantly contributed to inter-patient variability in warfarin dose requirements. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 15049511-0 2004 Inhibitory effects of the monoamine oxidase inhibitor tranylcypromine on the cytochrome P450 enzymes CYP2C19, CYP2C9, and CYP2D6. Tranylcypromine 54-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 15049511-2 2004 The inhibitory effects of tranylcypromine, a nonselective irreversible inhibitor of monoamine oxidase (MAO), on three cytochrome P450 (CYP) enzymes, namely CYP2C9, CYP2C19, and CYP2D6, have been evaluated in vitro. Tranylcypromine 26-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 15049511-9 2004 The results demonstrated that tranylcypromine is a competitive inhibitor of CYP2C19 (Ki = 32 microM) and CYP2D6 (Ki = 367 microM) and a noncompetitive inhibitor of CYP2C9 (Ki = 56 microM). Tranylcypromine 30-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 14718257-0 2004 5-Hydroxytryptamine is biotransformed by CYP2C9, 2C19 and 2B6 to hydroxylamine, which is converted into nitric oxide. Hydroxylamine 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 14718257-0 2004 5-Hydroxytryptamine is biotransformed by CYP2C9, 2C19 and 2B6 to hydroxylamine, which is converted into nitric oxide. Nitric Oxide 104-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 15499170-10 2004 On the other hand, bergamottin showed time-dependent decreases in IC(50) for CYP1A2, CYP 2C9, CYP 2C19, CYP 2D6 and CYP 3A4, suggesting that bergamottin is a quasi-irrversible or an irreversible inhibitor of the 5 CYP isoforms. bergamottin 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-92 15499170-10 2004 On the other hand, bergamottin showed time-dependent decreases in IC(50) for CYP1A2, CYP 2C9, CYP 2C19, CYP 2D6 and CYP 3A4, suggesting that bergamottin is a quasi-irrversible or an irreversible inhibitor of the 5 CYP isoforms. bergamottin 141-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-92 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. norfluoxetine 135-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. norfluoxetine 135-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 14726986-2 2004 CYP2C9 has also been shown to influence the metabolism of fluoxetine in vitro; however, this relationship has not been studied in humans. Fluoxetine 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. norfluoxetine 135-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 14726986-3 2004 The aim of the present study was to evaluate the influence of CYP2D6 and CYP2C9 genotypes on the plasma concentration of fluoxetine and norfluoxetine in psychiatric patients during steady-state conditions. Fluoxetine 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 14726986-11 2004 CONCLUSION: The present results show that CYP2D6 and potentially CYP2C9 genotypes seem to influence fluoxetine plasma concentration during steady-state conditions in patients. Fluoxetine 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 14726986-3 2004 The aim of the present study was to evaluate the influence of CYP2D6 and CYP2C9 genotypes on the plasma concentration of fluoxetine and norfluoxetine in psychiatric patients during steady-state conditions. norfluoxetine 136-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 14747429-9 2004 As fluconazole is a strong inhibitor of cytochrome P450 (CYP) 2C9, other CYP2C9 inhibitors are unlikely to affect lumiracoxib pharmacokinetics with clinical relevance, making dosage adjustment unnecessary. Fluconazole 3-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-65 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 14726986-9 2004 Among patients with two CYP2D6 active genes, the dose-corrected plasma concentrations of fluoxetine and active moiety (fluoxetine plus norfluoxetine) were significantly ( P<0.05) higher in the CYP2C9*1/*2 and CYP2C9*1/*3 genotype groups than in CYP2C9*1/*1. Fluoxetine 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 15066644-0 2004 Warfarin therapy is feasible in CYP2C9*3 homozygous patients. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 15066644-1 2004 Background: Patients carrying variant CYP2C9 alleles are prone to bleeding complications under standard warfarin treatment. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 14600250-8 2004 This is the first report to demonstrate that the nuclear receptor PXR mediates induction of CYP2C9 with rifampicin, phenobarbital, and hyperforin. hyperforin 135-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 14600250-0 2004 Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. Rifampin 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. Atomoxetine Hydrochloride 28-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 14600250-0 2004 Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. hyperforin 41-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14600250-0 2004 Induction of human CYP2C9 by rifampicin, hyperforin, and phenobarbital is mediated by the pregnane X receptor. Phenobarbital 57-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14600250-1 2004 Human CYP2C9 is important in the metabolism of numerous clinically used drugs such as the anticoagulant warfarin, the anticonvulsant phenytoin, antidiabetic drugs such as tolbutamide and glipizide, the hypertensive agent losartan, and numerous nonsteroidal anti-inflammatory drugs. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 14600250-1 2004 Human CYP2C9 is important in the metabolism of numerous clinically used drugs such as the anticoagulant warfarin, the anticonvulsant phenytoin, antidiabetic drugs such as tolbutamide and glipizide, the hypertensive agent losartan, and numerous nonsteroidal anti-inflammatory drugs. Phenytoin 133-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 14600250-1 2004 Human CYP2C9 is important in the metabolism of numerous clinically used drugs such as the anticoagulant warfarin, the anticonvulsant phenytoin, antidiabetic drugs such as tolbutamide and glipizide, the hypertensive agent losartan, and numerous nonsteroidal anti-inflammatory drugs. Tolbutamide 171-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 14600250-1 2004 Human CYP2C9 is important in the metabolism of numerous clinically used drugs such as the anticoagulant warfarin, the anticonvulsant phenytoin, antidiabetic drugs such as tolbutamide and glipizide, the hypertensive agent losartan, and numerous nonsteroidal anti-inflammatory drugs. Glipizide 187-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 14600250-1 2004 Human CYP2C9 is important in the metabolism of numerous clinically used drugs such as the anticoagulant warfarin, the anticonvulsant phenytoin, antidiabetic drugs such as tolbutamide and glipizide, the hypertensive agent losartan, and numerous nonsteroidal anti-inflammatory drugs. Losartan 221-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 14600250-2 2004 Several studies have reported that certain drugs such as rifampicin and phenobarbital induce CYP2C9, but the molecular basis for this induction remains unknown. Rifampin 57-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 14600250-2 2004 Several studies have reported that certain drugs such as rifampicin and phenobarbital induce CYP2C9, but the molecular basis for this induction remains unknown. Phenobarbital 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. Rifampin 131-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. hyperforin 143-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 14600250-3 2004 In the present study, we demonstrate that the human pregnane X receptor (hPXR) mediates induction of CYP2C9 by the prototype drugs rifampicin, hyperforin (found in St. John"s Wart), and phenobarbital. Phenobarbital 186-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 14600250-4 2004 Deletion and mutagenesis studies with luciferase reporter constructs showed that a functional PXR-responsive element located -1839/-1824 base pairs upstream from the translation start site was the primary binding site mediating the rifampicin induction of CYP2C9. Rifampin 232-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 256-262 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. N-desmethylatomoxetine 65-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 14600250-6 2004 Mutational analysis of 3- and 12-kilobase CYP2C9 promoter fragments indicated that this proximal binding site was essential for rifampicin inducibility, although a cooperative effect could be attributed to a second CAR-RE located at -2899/-2883. Rifampin 128-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 14600250-7 2004 In summary, we have demonstrated rifampicin induction of CYP2C9 promoter constructs that is consistent with the magnitude of induction of CYP2C9 protein and mRNA reported in vivo and in primary human hepatocytes, and we have identified the cis-element essential for this response. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 14600250-7 2004 In summary, we have demonstrated rifampicin induction of CYP2C9 promoter constructs that is consistent with the magnitude of induction of CYP2C9 protein and mRNA reported in vivo and in primary human hepatocytes, and we have identified the cis-element essential for this response. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 14600250-8 2004 This is the first report to demonstrate that the nuclear receptor PXR mediates induction of CYP2C9 with rifampicin, phenobarbital, and hyperforin. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 14610241-2 2004 Initially, the potential of atomoxetine and its two metabolites, N-desmethylatomoxetine and 4-hydroxyatomoxetine, to inhibit the metabolism of probe substrates for CYP1A2, CYP2C9, CYP2D6, and CYP3A was evaluated in human hepatic microsomes. 4-hydroxyatomoxetine 92-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 14600250-8 2004 This is the first report to demonstrate that the nuclear receptor PXR mediates induction of CYP2C9 with rifampicin, phenobarbital, and hyperforin. Phenobarbital 116-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 14985143-0 2004 CYP2C subfamily, primarily CYP2C9, catalyses the enantioselective demethylation of the endocrine disruptor pesticide methoxychlor in human liver microsomes: use of inhibitory monoclonal antibodies in P450 identification. Methoxychlor 117-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 14970301-0 2004 Warfarin and celecoxib interaction in the setting of cytochrome P450 (CYP2C9) polymorphism with bleeding complication. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 14970301-0 2004 Warfarin and celecoxib interaction in the setting of cytochrome P450 (CYP2C9) polymorphism with bleeding complication. Celecoxib 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 14970301-3 2004 As the CYP2C9 enzyme metabolises these drugs, it was determined whether variant alleles were responsible for altering warfarin handling. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 7-13 14662709-0 2004 Inhibition of cytochrome P450 2C9 improves endothelium-dependent, nitric oxide-mediated vasodilatation in patients with coronary artery disease. Nitric Oxide 66-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-33 14662709-10 2004 CONCLUSIONS: The CYP 2C9 inhibitor sulfaphenazole enhances endothelium-dependent vasodilator responses in patients with manifest coronary artery disease. Sulfaphenazole 35-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-24 14662709-11 2004 This effect seems to be related to an increase in the bioavailability of NO, probably as a consequence of an attenuated generation of reactive oxygen species by CYP 2C9 in endothelial cells. Reactive Oxygen Species 134-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-168 14659971-2 2004 Cytochrome P450 2C (CYP2C) plays an important role in phenytoin metabolism. Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-18 14659971-2 2004 Cytochrome P450 2C (CYP2C) plays an important role in phenytoin metabolism. Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-25 14659971-3 2004 Recently, single nucleotide polymorphisms in the coding region of CYP 2C influencing phenytoin metabolism were identified. Phenytoin 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 14659971-4 2004 The purpose of the present study was to see if CYP 2C polymorphisms might relate to the onset and severity of phenytoin-induced gingival overgrowth. Phenytoin 110-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 14628297-7 2004 At 10 microg/ml of methadone, CYP2C9 and CYP2D6 also generated EDDP, but in at least 10-fold lower quantities than CYP2B6. Methadone 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 14628297-7 2004 At 10 microg/ml of methadone, CYP2C9 and CYP2D6 also generated EDDP, but in at least 10-fold lower quantities than CYP2B6. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 63-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 15061384-3 2004 The aim of the study was to determine the frequency of the CYP2C9 polymorphism in a representative sample of the Croatian population (n = 177) and to assess the association between the CYP2C9 polymorphism and the warfarin dose in patients with thromboembolism (n = 181). Warfarin 213-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-191 15061384-16 2004 These preliminary results suggest a significant association of the CYP2C9 polymorphism with the warfarin dose and underline the importance of pre-therapeutic genotyping to identify the subjects likely to develop undesirable drug effects. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 14748619-7 2004 Nateglinide is extensively metabolised, primarily by cytochrome P450 2C9, and eliminated primarily by the kidney. Nateglinide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 15005635-0 2004 Influence of CYP2C9 and CYP2D6 polymorphisms on the pharmacokinetics of nateglinide in genotyped healthy volunteers. Nateglinide 72-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 15005635-2 2004 According to in vitro data, about 70% of nateglinide intrinsic clearance may be mediated by cytochrome P450 (CYP) 2C9 and a smaller fraction by CYP3A4 and CYP2D6. Nateglinide 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-117 15005635-7 2004 RESULTS: Significantly reduced oral nateglinide clearance was found in carriers of CYP2C9*3 alleles, (p < 0.01), whereas carriers of CYP2C9*2 alleles had kinetic parameters similar to those of carriers of the wild-type allele (p = nonsignificant). Nateglinide 36-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 15005635-10 2004 These differences in nateglinide kinetics due to CYP2C9 genotypes did not result in statistically significant differences in plasma glucose, insulin and glucagon. Nateglinide 21-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 15005635-11 2004 Pharmacokinetic-pharmacodynamic modelling revealed a minor effect of CYP2C9 genotype on insulin and glucose, and extrapolations indicated that carriers of the CYP2C9*3/*3 genotype may be at a slightly higher risk of hypoglycaemia compared with carriers of CYP2C9*1, particularly when taking nateglinide doses above 120 mg. Glucose 100-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 15005635-11 2004 Pharmacokinetic-pharmacodynamic modelling revealed a minor effect of CYP2C9 genotype on insulin and glucose, and extrapolations indicated that carriers of the CYP2C9*3/*3 genotype may be at a slightly higher risk of hypoglycaemia compared with carriers of CYP2C9*1, particularly when taking nateglinide doses above 120 mg. Glucose 100-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 14741071-1 2004 OBJECTIVE: The potential for a drug interaction was investigated between nateglinide, an oral antidiabetic agent, and acenocoumarol, an oral anticoagulant, as these drugs are primarily metabolized via CYP2C9. Nateglinide 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 15005635-11 2004 Pharmacokinetic-pharmacodynamic modelling revealed a minor effect of CYP2C9 genotype on insulin and glucose, and extrapolations indicated that carriers of the CYP2C9*3/*3 genotype may be at a slightly higher risk of hypoglycaemia compared with carriers of CYP2C9*1, particularly when taking nateglinide doses above 120 mg. Nateglinide 291-302 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 15005635-11 2004 Pharmacokinetic-pharmacodynamic modelling revealed a minor effect of CYP2C9 genotype on insulin and glucose, and extrapolations indicated that carriers of the CYP2C9*3/*3 genotype may be at a slightly higher risk of hypoglycaemia compared with carriers of CYP2C9*1, particularly when taking nateglinide doses above 120 mg. Nateglinide 291-302 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 15005635-12 2004 CONCLUSION: The effect of CYP2C9 polymorphisms on nateglinide kinetics may cause a slightly increased risk for hypoglycaemia, which may become relevant in diabetic patients. Nateglinide 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 15509184-13 2004 In vitro data suggest that (R)-bicalutamide has the potential to inhibit CYP3A4 and, to a lesser extent, CYP2C9, 2C19 and 2D6. (R)-Bicalutamide 27-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 15568889-16 2004 Bosentan decreases exposure to ciclosporin, glibenclamide, simvastatin (and beta-hydroxyacid simvastatin) and (R)- and (S)-warfarin by up to 50% because of induction of CYP3A4 and/or CYP2C9. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 15568889-20 2004 The possibility of reduced efficacy of CYP2C9 and 3A4 substrates should be considered when coadministered with bosentan. Bosentan 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 14971821-4 2004 The patient was homozygous for the rare CYP2C9*3 allele, which determines the slowest metabolic rate for CYP2C9 enzymatic activity, that is probably involved in the metabolism of leflunomide. Leflunomide 179-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 14971821-4 2004 The patient was homozygous for the rare CYP2C9*3 allele, which determines the slowest metabolic rate for CYP2C9 enzymatic activity, that is probably involved in the metabolism of leflunomide. Leflunomide 179-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 14709627-5 2004 The reference inhibitor ketoconazole was less selective and exhibited potent inhibition (IC(50) values <10 microM) for CYP1A1, CYP1B1, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP4F2, and CYP4F12. Ketoconazole 24-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 14709614-0 2004 Catalytic roles of CYP2C9 and its variants (CYP2C9*2 and CYP2C9*3) in lornoxicam 5"-hydroxylation. lornoxicam 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14709614-0 2004 Catalytic roles of CYP2C9 and its variants (CYP2C9*2 and CYP2C9*3) in lornoxicam 5"-hydroxylation. lornoxicam 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 14709614-0 2004 Catalytic roles of CYP2C9 and its variants (CYP2C9*2 and CYP2C9*3) in lornoxicam 5"-hydroxylation. lornoxicam 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 14563790-5 2004 Clopidogrel also inhibited CYP2C9, and ticlopidine also inhibited CYP1A2, with lower potency. Clopidogrel 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 14709614-1 2004 The effects of allelic variants of CYP2C9 (CYP2C9*2 and CYP2C9*3) on lornoxicam 5"-hydroxylation were studied using the corresponding variant protein expressed in baculovirus-infected insect cells and human liver microsomes of known genotypes of CYP2C9. lornoxicam 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14709614-1 2004 The effects of allelic variants of CYP2C9 (CYP2C9*2 and CYP2C9*3) on lornoxicam 5"-hydroxylation were studied using the corresponding variant protein expressed in baculovirus-infected insect cells and human liver microsomes of known genotypes of CYP2C9. lornoxicam 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 14709614-1 2004 The effects of allelic variants of CYP2C9 (CYP2C9*2 and CYP2C9*3) on lornoxicam 5"-hydroxylation were studied using the corresponding variant protein expressed in baculovirus-infected insect cells and human liver microsomes of known genotypes of CYP2C9. lornoxicam 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 14709614-1 2004 The effects of allelic variants of CYP2C9 (CYP2C9*2 and CYP2C9*3) on lornoxicam 5"-hydroxylation were studied using the corresponding variant protein expressed in baculovirus-infected insect cells and human liver microsomes of known genotypes of CYP2C9. lornoxicam 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 14709614-2 2004 The results of the baculovirus expression system showed that CYP2C9.3 gives higher K(m) and lower V(max) values for lornoxicam 5"-hydroxylation than does CYP2C9.1. lornoxicam 116-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. lornoxicam 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. lornoxicam 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. lornoxicam 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. ile359leu 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. ile359leu 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 14709614-8 2004 In conclusion, this study showed that lornoxicam 5"-hydroxylation via CYP2C9 was markedly decreased by the substitution of Ile359Leu (CYP2C9.3), whereas the effect of the substitution of Arg144Cys (CYP2C9.2) was nonexistent or negligible. ile359leu 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 14709614-9 2004 Additional in vivo studies are required to confirm that individuals with homologous CYP2C9*3 allele exhibit impaired lornoxicam clearance. lornoxicam 117-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14676821-2 2004 Warfarin is metabolised by cytochrome P450 enzymes CYP2C9, CYP1A2 and CYP3A. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 14676821-4 2004 Genetic variability in CYP2C9, CYP3A5 and ABCB1 was analysed in 201 stable warfarin-treated patients using solid-phase minisequencing, pyrosequencing and SNaPshot. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 14676821-5 2004 CYP2C9 variants, age, weight, concurrent drug treatment and indication for treatment significantly influenced warfarin dosing in these patients, explaining 29% of the variation in dose. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15128047-1 2004 CYP2C9 catalyses the biotransformation of the oral anticoagulants S-warfarin and R- and S-acenocoumarol. Sulfur 66-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15128047-1 2004 CYP2C9 catalyses the biotransformation of the oral anticoagulants S-warfarin and R- and S-acenocoumarol. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15128047-1 2004 CYP2C9 catalyses the biotransformation of the oral anticoagulants S-warfarin and R- and S-acenocoumarol. r- and s-acenocoumarol 81-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15128047-2 2004 According to data obtained in vitro, phenprocoumon is also metabolized by CYP2C9 but the impact of the CYP2C9 polymorphism on phenprocoumon pharmacokinetics has not been studied. Phenprocoumon 37-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 15128047-3 2004 Twenty-six healthy heterozygous and homozygous carriers of the CYP2C9 alleles *1 (wild-type), *2 (Arg144Cys), and *3 (Ile359Leu) received a single oral dose of 12 mg of racemic phenprocoumon. ile359leu 118-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15128047-3 2004 Twenty-six healthy heterozygous and homozygous carriers of the CYP2C9 alleles *1 (wild-type), *2 (Arg144Cys), and *3 (Ile359Leu) received a single oral dose of 12 mg of racemic phenprocoumon. Phenprocoumon 177-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 15128047-7 2004 Plasma and urine concentrations of 4"-, 6- and 7-hydroxyphenprocoumon were significantly lower in homozygous carriers of the CYP2C9*2 and *3 variants compared to CYP2C9*1/*1. 4"-, 6- and 7-hydroxyphenprocoumon 35-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 15128047-8 2004 Carriers of CYP2C9*3/*3 had a median AUC of (R,S) 7-OH-phenprocoumon of only approximately 25% compared to the wild-type genotype. (r,s) 7-oh-phenprocoumon 44-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 15128047-9 2004 The AUC of (R,S) 6-OH-phenprocoumon was only approximately 50% in CYP2C9*3/*3 compared to the homozygous wild-type genotype. (r,s) 6-oh-phenprocoumon 11-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 15128048-0 2004 The risk of overanticoagulation in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Acenocoumarol 97-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 15128048-0 2004 The risk of overanticoagulation in patients with cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenprocoumon. Acenocoumarol 97-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 15128048-1 2004 Cytochrome P4502C9 (CYP2C9) is the main enzyme implicated in coumarin anticoagulant metabolism. coumarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-18 15128048-1 2004 Cytochrome P4502C9 (CYP2C9) is the main enzyme implicated in coumarin anticoagulant metabolism. coumarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 15128048-2 2004 The variant alleles CYP2C9*2 and CYP2C9*3 are associated with an increased response to warfarin. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 15128048-2 2004 The variant alleles CYP2C9*2 and CYP2C9*3 are associated with an increased response to warfarin. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 15128048-4 2004 We investigated CYP2C9 polymorphisms in relation to the international normalized ratio (INR) during the first 6 weeks of treatment and its effect on the maintenance dose in a cohort of 1124 patients from the Rotterdam Study who were treated with acenocoumarol or phenprocoumon. Acenocoumarol 246-259 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 15128048-4 2004 We investigated CYP2C9 polymorphisms in relation to the international normalized ratio (INR) during the first 6 weeks of treatment and its effect on the maintenance dose in a cohort of 1124 patients from the Rotterdam Study who were treated with acenocoumarol or phenprocoumon. Phenprocoumon 263-276 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 15128048-9 2004 Individuals with one or more CYP2C9*2 or CYP2C9*3 allele(s) require a significantly lower dose of acenocoumarol compared to wild-type patients. Acenocoumarol 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 15128048-9 2004 Individuals with one or more CYP2C9*2 or CYP2C9*3 allele(s) require a significantly lower dose of acenocoumarol compared to wild-type patients. Acenocoumarol 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 14691573-6 2004 Advanced age, lower body surface area (BSA), and the presence of cytochrome P450 2C9 *2 or *3 single nucleotide polymorphisms were strongly associated (P < 0.001) with lower warfarin dose: the maintenance dose decreased by 8% per decade of age, by 13% per standard deviation decrease in BSA, by 19% per 2C9*2 allele, and by 30% per 2C9*3 allele. Warfarin 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-84 14676821-0 2004 Warfarin sensitivity related to CYP2C9, CYP3A5, ABCB1 (MDR1) and other factors. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 14691574-4 2004 We also determined the role of cytochrome P450 2C9 (CYP2C9) polymorphism on coumarin dosage and INR in NSAID users. coumarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-50 14691574-4 2004 We also determined the role of cytochrome P450 2C9 (CYP2C9) polymorphism on coumarin dosage and INR in NSAID users. coumarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 14642736-7 2003 Incubations of tamoxifen with recombinant human cytochrome P-450s (CYPs) found that CYP2C9 and CYP2D6 produced all three of the above tamoxifen metabolites, while CYP1A1 and CYP3A4 catalyzed the formation of alpha-hydroxytamoxifen and N-desmethyltamoxifen, and CYP1A2 and CYP1B1 only formed the alpha-hydroxy metabolite. Tamoxifen 15-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 15199661-5 2004 Fluvoxamine is a potent inhibitor of CYP1A2 and CYP2C19 and a moderate inhibitor of CYP2C9. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14614357-0 2003 Early overanticoagulation with acenocoumarol due to a genetic polymorphism of cytochrome P450 CYP2C9. Acenocoumarol 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 14642736-7 2003 Incubations of tamoxifen with recombinant human cytochrome P-450s (CYPs) found that CYP2C9 and CYP2D6 produced all three of the above tamoxifen metabolites, while CYP1A1 and CYP3A4 catalyzed the formation of alpha-hydroxytamoxifen and N-desmethyltamoxifen, and CYP1A2 and CYP1B1 only formed the alpha-hydroxy metabolite. Tamoxifen 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14614357-3 2003 This case illustrates the role of the *3 polymorphism of the cytochrome P450 CYP2C9 as an independent risk factor modulating the sensitivity of patients to the anticoagulant effect of acenocoumarol. Acenocoumarol 184-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 14616412-1 2003 AIMS: Voriconazole is a new triazole antifungal agent, and is metabolized by the cytochrome P450 isoenzymes CYP2C9, CYP2C19, and, to a lesser extent, by CYP3A4. Voriconazole 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 14642736-7 2003 Incubations of tamoxifen with recombinant human cytochrome P-450s (CYPs) found that CYP2C9 and CYP2D6 produced all three of the above tamoxifen metabolites, while CYP1A1 and CYP3A4 catalyzed the formation of alpha-hydroxytamoxifen and N-desmethyltamoxifen, and CYP1A2 and CYP1B1 only formed the alpha-hydroxy metabolite. alpha-hydroxytamoxifen 208-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14642736-7 2003 Incubations of tamoxifen with recombinant human cytochrome P-450s (CYPs) found that CYP2C9 and CYP2D6 produced all three of the above tamoxifen metabolites, while CYP1A1 and CYP3A4 catalyzed the formation of alpha-hydroxytamoxifen and N-desmethyltamoxifen, and CYP1A2 and CYP1B1 only formed the alpha-hydroxy metabolite. N-desmethyltamoxifen 235-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 14616412-1 2003 AIMS: Voriconazole is a new triazole antifungal agent, and is metabolized by the cytochrome P450 isoenzymes CYP2C9, CYP2C19, and, to a lesser extent, by CYP3A4. Triazoles 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 14616412-2 2003 Phenytoin is an inducer of CYP3A4 activity, and a substrate and inducer of CYP2C9 and CYP2C19. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 14616414-1 2003 AIMS: Voriconazole, a new triazole antifungal agent, is metabolized mainly by cytochrome P450s CYP2C19 and CYP2C9, and also by CYP3A4. Voriconazole 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 14616414-1 2003 AIMS: Voriconazole, a new triazole antifungal agent, is metabolized mainly by cytochrome P450s CYP2C19 and CYP2C9, and also by CYP3A4. Triazoles 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 14616415-1 2003 AIMS: Voriconazole, a novel triazole antifungal agent, is metabolized by the cytochrome P450 isoenzymes CYP2C19, CYP2C9, and to a lesser extent by CYP3A4. Voriconazole 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 14616416-1 2003 AIMS: Voriconazole is a new triazole antifungal agent, and is metabolized by the cytochrome P450 isoenzymes CYP2C9, CYP2C19 and to a lesser extent by CYP3A4. Voriconazole 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 14616416-1 2003 AIMS: Voriconazole is a new triazole antifungal agent, and is metabolized by the cytochrome P450 isoenzymes CYP2C9, CYP2C19 and to a lesser extent by CYP3A4. Triazoles 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 14703066-9 2003 CYP2C9 is involved in the conversion of estrone sulfate to the 16-hydroxy sulfate metabolite. estrone sulfate 40-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 14703066-9 2003 CYP2C9 is involved in the conversion of estrone sulfate to the 16-hydroxy sulfate metabolite. 16-hydroxy sulfate 63-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 14575648-4 2003 Among these are metabolic factors (bioactivation by hCYP2C9 or hCYP3A4 to thiol-reactive quinone imines, activation by hUGT2B7 to protein-reactive acyl glucuronides and iso-glucuronides, and 4"-hydroxylation secondary to diclofenac glucuronidation), as well as kinetic factors (Mrp2-mediated concentrative transport of diclofenac metabolites into bile). Sulfhydryl Compounds 74-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-59 14684755-0 2003 Involvement of CYP 2C9 in mediating the proinflammatory effects of linoleic acid in vascular endothelial cells. Linoleic Acid 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-22 14684755-3 2003 CYP 2C9 is involved in linoleic acid epoxygenation, and the major product of this reaction is leukotoxin (LTX). Linoleic Acid 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 14684755-5 2003 METHODS: The effect of linoleic acid on CYP 2C9 gene expression was studied by RT-PCR. Linoleic Acid 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-47 14684755-8 2003 RESULTS: Linoleic acid treatment for six hours increased the expression of CYP 2C9 in endothelial cells. Linoleic Acid 9-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-82 14684755-9 2003 Linoleic acid-mediated increase in oxidative stress and activation of AP-1 were blocked by sulfaphenazole, a specific inhibitor of CYP 2C9. Linoleic Acid 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-138 14684755-9 2003 Linoleic acid-mediated increase in oxidative stress and activation of AP-1 were blocked by sulfaphenazole, a specific inhibitor of CYP 2C9. Sulfaphenazole 91-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-138 14684755-11 2003 CONCLUSION: Our data show that CYP 2C9 plays a key role in linoleic acid-induced oxidative stress and subsequent proinflammatory events in vascular endothelial cells by possibly causing superoxide generation through uncoupling processes. Linoleic Acid 59-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-38 14684755-11 2003 CONCLUSION: Our data show that CYP 2C9 plays a key role in linoleic acid-induced oxidative stress and subsequent proinflammatory events in vascular endothelial cells by possibly causing superoxide generation through uncoupling processes. Superoxides 186-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-38 14651673-0 2003 Involvement of cytochrome P450 2C9, 2E1 and 3A4 in trimethadione N-demethylation in human microsomes. Trimethadione 51-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-34 14651673-3 2003 In this study, we determined the percentage contribution of the three CYPs (CYP2C9, CYP2E1 and CYP3A4) to TMO N-demethylation. Nitrogen 110-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 14651673-9 2003 CONCLUSION: TMO is metabolized not only by CYP2E1 but also CYP3A4 and CYP2C9. Trimethadione 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). 7-methoxy-4-trifluoromethylcoumarin 51-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). Amiodarone 143-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). Niclosamide 155-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). Triiodothyronine 168-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). meclofenemate 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). zafirlukast 197-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). estropipate 210-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14557374-3 2003 Several potent heteroactivators of CYP2C9-mediated 7-methoxy-4-trifluoromethyl-coumarin metabolism are marketed drugs or endogenous compounds (amiodarone, niclosamide, liothyronine, meclofenemate, zafirlukast, estropipate, and dichlorphenamide, yielding 150% control reaction velocity at 0.04, 0.09, 0.5, 1, 1.2, 1.5, and 2.5 microM, respectively). Dichlorphenamide 227-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14576103-4 2003 The mechanism of the drug interaction reported between chloramphenicol and phenytoin, which results in the elevation of plasma phenytoin concentrations, is clinically assumed to result from the inhibition of CYP2C9 by chloramphenicol. Chloramphenicol 55-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 14576103-4 2003 The mechanism of the drug interaction reported between chloramphenicol and phenytoin, which results in the elevation of plasma phenytoin concentrations, is clinically assumed to result from the inhibition of CYP2C9 by chloramphenicol. Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 14576103-4 2003 The mechanism of the drug interaction reported between chloramphenicol and phenytoin, which results in the elevation of plasma phenytoin concentrations, is clinically assumed to result from the inhibition of CYP2C9 by chloramphenicol. Phenytoin 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 14576103-4 2003 The mechanism of the drug interaction reported between chloramphenicol and phenytoin, which results in the elevation of plasma phenytoin concentrations, is clinically assumed to result from the inhibition of CYP2C9 by chloramphenicol. Chloramphenicol 218-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 14661864-0 2003 Polymorphism in CYP2C9 as a non-critical factor of warfarin dosage adjustment in Korean patients. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 14661864-1 2003 Cytochrome P4502C9(CYP2C9) is largely responsible for terminating anticoagulant effect by hydroxylation of S-warfarin to inactive metabolites. Warfarin 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 14661864-3 2003 The aim of this study was to assess relationship between requirement of warfarin dose and polymorphism in CYP2C9 in Korean population. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 14586384-2 2003 Data suggest that warfarin can be used as a safe and accurate biomarker for CYP2C9, and if warfarin is administered with vitamin K, the pharmacodynamic effect is ablated. Warfarin 18-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 14586391-6 2003 These results suggest that the CYP2C9*1/*3 genotype could be associated with decreased clearance and increased plasma concentration of candesartan, potentially enhancing its hypotensive effect. candesartan 135-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 14678248-8 2003 Cytochrome P450 (CYP) isoform-specific chemical and monoclonal antibody inhibition studies have demonstrated that CYP2B6, CYP2C9, CYP2D6 and CYP3A4 all mediate the formation of Z-4-OH-tam. z-4-oh-tam 177-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 14678248-10 2003 Significant associations between the percentage inhibition of Z-4-OH-tam by CYP isoform-specific inhibitors and the rate of metabolism of CYP isoform-specific index reactions and between individual expression of CYP2B6, CYP2C9 and CYP2D6 and Z-4-OH-tam formation rates indicate predominant roles for these isoforms in this pathway. z-4-oh-tam 62-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 14678248-12 2003 Genotyping of patients with regards to CYP2B6, CYP2C9 and CYP2D6 may play a role in prediction of Z-4-OH-tam formation and, consequently, ultimate therapeutic benefit of tamoxifen treatment. z-4-oh-tam 98-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 14678248-12 2003 Genotyping of patients with regards to CYP2B6, CYP2C9 and CYP2D6 may play a role in prediction of Z-4-OH-tam formation and, consequently, ultimate therapeutic benefit of tamoxifen treatment. Tamoxifen 170-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 14504849-0 2003 Effect of the single CYP2C9*3 allele on pharmacokinetics and pharmacodynamics of losartan in healthy Japanese subjects. Losartan 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 14504849-1 2003 OBJECTIVE: Losartan is metabolized to the active carboxylic acid metabolite EXP3174 by CYP2C9. Losartan 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 14504849-1 2003 OBJECTIVE: Losartan is metabolized to the active carboxylic acid metabolite EXP3174 by CYP2C9. Carboxylic Acids 49-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 14504849-2 2003 In this study, we determined the effects of the single CYP2C9*3 variant on the pharmacokinetics and pharmacodynamics of losartan. Losartan 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 14504849-11 2003 CONCLUSION: The single CYP2C9*3 variant reduces the metabolism of losartan and its hypotensive effect. Losartan 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 14641553-2 2003 Of special interest are those with a narrow therapeutic index, such as S-warfarin, tolbutamide and phenytoin, where impairment in CYP2C9 metabolic activity might cause difficulties in dose adjustment as well as toxicity. Sulfur 71-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 14641553-2 2003 Of special interest are those with a narrow therapeutic index, such as S-warfarin, tolbutamide and phenytoin, where impairment in CYP2C9 metabolic activity might cause difficulties in dose adjustment as well as toxicity. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 14641553-2 2003 Of special interest are those with a narrow therapeutic index, such as S-warfarin, tolbutamide and phenytoin, where impairment in CYP2C9 metabolic activity might cause difficulties in dose adjustment as well as toxicity. Tolbutamide 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 14641553-2 2003 Of special interest are those with a narrow therapeutic index, such as S-warfarin, tolbutamide and phenytoin, where impairment in CYP2C9 metabolic activity might cause difficulties in dose adjustment as well as toxicity. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 14641553-8 2003 Furthermore in Japanese a CYP2C9 promotor variant of four linked SNPs was correlated with reduced intrinsic clearance of phenytoin in vitro. Phenytoin 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 12826020-2 2003 The cytochrome P450 (CYP) 2C9 inhibitor sulphaphenazole was recently identified as a potent inhibitor of NO- and prostacyclin (PGI2)-independent relaxation in porcine coronary arteries. Sulfaphenazole 40-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-29 12826020-2 2003 The cytochrome P450 (CYP) 2C9 inhibitor sulphaphenazole was recently identified as a potent inhibitor of NO- and prostacyclin (PGI2)-independent relaxation in porcine coronary arteries. Epoprostenol 113-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-29 12826020-2 2003 The cytochrome P450 (CYP) 2C9 inhibitor sulphaphenazole was recently identified as a potent inhibitor of NO- and prostacyclin (PGI2)-independent relaxation in porcine coronary arteries. Epoprostenol 127-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-29 14520122-0 2003 Effects of polymorphisms in CYP2D6, CYP2C9, and CYP2C19 on trimipramine pharmacokinetics. Trimipramine 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 14520122-2 2003 A single oral dose of 75 mg trimipramine was given to 42 healthy volunteers selected according to their CYP2D6, CYP2C19, and CYP2C9 genotypes. Trimipramine 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 14520122-8 2003 The desmethyltrimipramine area under the concentration-time curve was 40-fold greater in CYP2D6 PMs than in the reference group (1.7 vs. 0.04 mg/L x h in EMs), but below the quantification limit in most carriers of deficiencies of CYP2C19 or CYP2C9. desmethyltrimipramine 4-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 242-248 15647404-3 2005 Anticoagulant response is also influenced by a number of drugs that induce or inhibit warfarin metabolism, as well as by genetic polymorphisms that may modulate expression or activity of CYP2C9, the isoform mediating clearance of S-warfarin. Sulfur 230-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 15647404-3 2005 Anticoagulant response is also influenced by a number of drugs that induce or inhibit warfarin metabolism, as well as by genetic polymorphisms that may modulate expression or activity of CYP2C9, the isoform mediating clearance of S-warfarin. Warfarin 232-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 15647404-5 2005 St. John"s wort and possibly some ginseng formulations may have the potential to diminish warfarin anticoagulation, apparently by inducing CYP2C9 activity. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 14534520-8 2003 CONCLUSION: The effect of rifampin on the pharmacokinetics and pharmacodynamics of gliclazide suggests that rifampin affects the disposition of gliclazide in humans, possibly by the induction of cytochrome P450 2C9. Gliclazide 83-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-214 14534520-8 2003 CONCLUSION: The effect of rifampin on the pharmacokinetics and pharmacodynamics of gliclazide suggests that rifampin affects the disposition of gliclazide in humans, possibly by the induction of cytochrome P450 2C9. Rifampin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-214 14534520-8 2003 CONCLUSION: The effect of rifampin on the pharmacokinetics and pharmacodynamics of gliclazide suggests that rifampin affects the disposition of gliclazide in humans, possibly by the induction of cytochrome P450 2C9. Gliclazide 144-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-214 14521618-0 2003 Warfarin and acenocoumarol dose requirements according to CYP2C9 genotyping in North-Italian patients. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 14521618-0 2003 Warfarin and acenocoumarol dose requirements according to CYP2C9 genotyping in North-Italian patients. Acenocoumarol 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 12904931-4 2003 METHODS: The inhibitory effect of 5-FU on CYP1A2, CYP2C9, CYP2C19, CYP2C8, CYP2E1, CYP2D6, and CYP3A4 activities was examined with specific probe drugs in human liver microsomes. Fluorouracil 34-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 12972955-6 2003 The CLint for the formation of D-703 from verapamil (marker of CYP2C8 activity) was 7.6-fold higher (P < 0.001) and that for the diclofenac 4"-hydroxylation (marker of CYP2C9 activity) was 6.1-fold higher (P < 0.001) in the liver than in the intestine. D 703 31-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 12867493-2 2003 Inhibition of the CYP2C9-dependent formation of (S)-7-hydroxy-warfarin from (S)-warfarin was measured in seven healthy subjects at three different doses of fluconazole. 7-hydroxywarfarin 48-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 12920163-1 2003 The inhibitory effect of glyburide [International Nonproprietary Name (INN), glibenclamide] on CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4 activities was evaluated using pooled human liver microsomes. Glyburide 25-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 12920163-2 2003 Glyburide strongly inhibited CYP2C9-catalyzed S-warfarin and phenytoin metabolism in a competitive manner, with Ki (IC50) values of 2.4 (11.3) microM and 3.1 (9.4) microM, respectively. Glyburide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 12920163-2 2003 Glyburide strongly inhibited CYP2C9-catalyzed S-warfarin and phenytoin metabolism in a competitive manner, with Ki (IC50) values of 2.4 (11.3) microM and 3.1 (9.4) microM, respectively. Sulfur 46-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 12920163-2 2003 Glyburide strongly inhibited CYP2C9-catalyzed S-warfarin and phenytoin metabolism in a competitive manner, with Ki (IC50) values of 2.4 (11.3) microM and 3.1 (9.4) microM, respectively. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 12920163-2 2003 Glyburide strongly inhibited CYP2C9-catalyzed S-warfarin and phenytoin metabolism in a competitive manner, with Ki (IC50) values of 2.4 (11.3) microM and 3.1 (9.4) microM, respectively. Phenytoin 61-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 12920163-5 2003 In summary, glyburide showed potent inhibition on CYP2C9 and weak inhibition on CYP3A4, whereas it had minimal or no inhibitory effect on the other cytochromes p450 examined. Glyburide 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 12920163-6 2003 It is anticipated that clinically significant drug-drug interactions will ensue when glyburide is coadministered with agents that are cleared primarily by the CYP2C9-mediated pathway and those with narrow therapeutic ranges. Glyburide 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 12867493-2 2003 Inhibition of the CYP2C9-dependent formation of (S)-7-hydroxy-warfarin from (S)-warfarin was measured in seven healthy subjects at three different doses of fluconazole. Warfarin 76-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 12867493-2 2003 Inhibition of the CYP2C9-dependent formation of (S)-7-hydroxy-warfarin from (S)-warfarin was measured in seven healthy subjects at three different doses of fluconazole. Fluconazole 156-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 12900870-2 2003 Mephenytoin (MEPH), dextromethorphan, diclofenac, caffeine, and methadone (MET) were successfully applied as test substrates for CYP2C19, CYP2D6*1, CYP2C9*1, CYP1A2, and CYP3A4, respectively. Mephenytoin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12900870-2 2003 Mephenytoin (MEPH), dextromethorphan, diclofenac, caffeine, and methadone (MET) were successfully applied as test substrates for CYP2C19, CYP2D6*1, CYP2C9*1, CYP1A2, and CYP3A4, respectively. Dextromethorphan 20-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12900870-2 2003 Mephenytoin (MEPH), dextromethorphan, diclofenac, caffeine, and methadone (MET) were successfully applied as test substrates for CYP2C19, CYP2D6*1, CYP2C9*1, CYP1A2, and CYP3A4, respectively. Diclofenac 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12900870-2 2003 Mephenytoin (MEPH), dextromethorphan, diclofenac, caffeine, and methadone (MET) were successfully applied as test substrates for CYP2C19, CYP2D6*1, CYP2C9*1, CYP1A2, and CYP3A4, respectively. Methadone 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12900870-2 2003 Mephenytoin (MEPH), dextromethorphan, diclofenac, caffeine, and methadone (MET) were successfully applied as test substrates for CYP2C19, CYP2D6*1, CYP2C9*1, CYP1A2, and CYP3A4, respectively. Methadone 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12900870-7 2003 CYP2C9 and CYP2C19 provided enhanced formation of R-EDDP and CYP2D6 incubation resulted in the preferential conversion to S-EDDP. r-eddp 50-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 12900870-7 2003 CYP2C9 and CYP2C19 provided enhanced formation of R-EDDP and CYP2D6 incubation resulted in the preferential conversion to S-EDDP. 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 122-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15199455-3 2003 The most important of these is CYP2C9, which 7-hydroxylates S-warfarin. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12871048-2 2003 As a result, the metabolism of diclofenac in humans partitions between acyl glucuronidation and phenyl hydroxylation, with the former reaction catalyzed primarily by uridine 5"-diphosphoglucuronosyl transferase 2B7 while the latter is catalyzed by cytochrome P450 (CYP)2C9 and 3A4. Diclofenac 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 248-272 12871048-4 2003 The 4"-hydroxylation of the drug appears to represent a feature reaction for CYP2C9 catalysis, and this regioselective oxidation is presumably dictated by interactions of the carboxylate moiety of the substrate with a putative cationic residue of the enzyme. carboxylate 175-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 14760217-1 2003 Warfarin and other coumarins are metabolized by the cytochrome P450 2C9 complex. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 14760217-1 2003 Warfarin and other coumarins are metabolized by the cytochrome P450 2C9 complex. Coumarins 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 12893985-0 2003 Influence of CYP2C9 genetic polymorphisms on pharmacokinetics of celecoxib and its metabolites. Celecoxib 65-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12893985-1 2003 In-vitro data indicate major effects of the genetically polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) on the pharmacokinetics of celecoxib, a nonsteroidal anti-inflammatory drug acting as selective cyclooxygenase-2 inhibitor. Celecoxib 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-94 12893985-1 2003 In-vitro data indicate major effects of the genetically polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) on the pharmacokinetics of celecoxib, a nonsteroidal anti-inflammatory drug acting as selective cyclooxygenase-2 inhibitor. Celecoxib 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 12893985-2 2003 Human studies report decreased clearance in heterozygous carriers of the CYP2C9 variant Ile359Leu (*3), but results appeared controversial and only data on single subjects carrying the homozygous CYP2C9*3/*3 genotype have been published. ile359leu 88-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 12891229-0 2003 Influence of CYP2C9 polymorphisms on the pharmacokinetics and cholesterol-lowering activity of (-)-3S,5R-fluvastatin and (+)-3R,5S-fluvastatin in healthy volunteers. Cholesterol 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12891229-0 2003 Influence of CYP2C9 polymorphisms on the pharmacokinetics and cholesterol-lowering activity of (-)-3S,5R-fluvastatin and (+)-3R,5S-fluvastatin in healthy volunteers. (-)-3s,5r-fluvastatin 95-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12891229-2 2003 The consequences of CYP2C9 genetic polymorphisms on fluvastatin pharmacokinetics and on its efficacy have not been investigated in humans thus far. Fluvastatin 52-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 12891229-9 2003 Mean (and SD) values for area under the curve of the active (+)-3R,5S-fluvastatin in carriers of the genotype CYP2C9*1/*1, *1/*3, and *3/*3 were 173 (85) micro g. L(-1). 5s-fluvastatin 67-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 12891229-19 2003 CONCLUSIONS: The pharmacokinetics of both enantiomers of fluvastatin depended on the CYP2C9 genotype, with a 3-fold group mean difference in the active enantiomer and even greater differences in the inactive enantiomer, but differences in plasma concentrations were not reflected in cholesterol lowering after 14 days of fluvastatin intake in healthy volunteers. Fluvastatin 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 12891229-19 2003 CONCLUSIONS: The pharmacokinetics of both enantiomers of fluvastatin depended on the CYP2C9 genotype, with a 3-fold group mean difference in the active enantiomer and even greater differences in the inactive enantiomer, but differences in plasma concentrations were not reflected in cholesterol lowering after 14 days of fluvastatin intake in healthy volunteers. Cholesterol 283-294 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 12891229-19 2003 CONCLUSIONS: The pharmacokinetics of both enantiomers of fluvastatin depended on the CYP2C9 genotype, with a 3-fold group mean difference in the active enantiomer and even greater differences in the inactive enantiomer, but differences in plasma concentrations were not reflected in cholesterol lowering after 14 days of fluvastatin intake in healthy volunteers. Fluvastatin 321-332 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 15199455-4 2003 In clinical studies, possession of the CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, has been associated with a significant decrease in mean warfarin dose requirement in at least eight studies. Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 15199455-4 2003 In clinical studies, possession of the CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, has been associated with a significant decrease in mean warfarin dose requirement in at least eight studies. Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 15199455-7 2003 The molecular basis of warfarin resistance remains unclear but could be due to unusually high CYP2C9 activity (pharmacokinetic resistance) or to abnormal vitamin K epoxide reductase (pharmacodynamic resistance). Warfarin 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 15199455-8 2003 There is less information available on genetic factors affecting other anticoagulants, but the CYP2C9 genotype is also relevant to acenocoumarol dose. Acenocoumarol 131-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 12861225-0 2003 Crystal structure of human cytochrome P450 2C9 with bound warfarin. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-46 12899669-0 2003 A screening study on the liability of eight different female sex steroids to inhibit CYP2C9, 2C19 and 3A4 activities in human liver microsomes. Steroids 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 12899669-3 2003 Aethinyloestradiol was found to be a potent inhibitor (55% mean inhibition; 95% CI 32% to 79%) of losartan oxidation (CYP2C9) and R-omeprazole 5-hydroxylation (70%; 63% to 77%) (CYP2C19), while it had little effect on R-omeprazole sulphoxidation (CYP3A4) activity. aethinyloestradiol 0-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 12899669-3 2003 Aethinyloestradiol was found to be a potent inhibitor (55% mean inhibition; 95% CI 32% to 79%) of losartan oxidation (CYP2C9) and R-omeprazole 5-hydroxylation (70%; 63% to 77%) (CYP2C19), while it had little effect on R-omeprazole sulphoxidation (CYP3A4) activity. Losartan 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 12899669-5 2003 Of the progestin hormones studied, gestodene and 3-ketodesogestrel were potent inhibitors of CYP2C19 (57%; 47% to 67% and 51%; 29% to 45%) and CYP3A4 (45%; 30% to 59% and 40%; 19% to 62%), but had little effect on the CYP2C9 activity. Gestodene 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 12899669-5 2003 Of the progestin hormones studied, gestodene and 3-ketodesogestrel were potent inhibitors of CYP2C19 (57%; 47% to 67% and 51%; 29% to 45%) and CYP3A4 (45%; 30% to 59% and 40%; 19% to 62%), but had little effect on the CYP2C9 activity. etonogestrel 49-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 12899669-6 2003 In addition, medroxyprogesterone acetate was found to inhibit CYP2C9 (55%; 45% to 65%), while not having significant effect on 2C19 or 3A4. Medroxyprogesterone Acetate 13-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 12899669-7 2003 In conclusion, the liability of clinically used female sex steroids to inhibit CYP2C9, 2C19 and 3A4 activities in human liver microsomes is very distinctive and these differences among both the oestrogen and progestin hormones may, at least in part, explain the variable results from clinical trials examining inhibitory effects of hormone replacement therapy and oral contraceptives on drug metabolism. Steroids 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 12861225-6 2003 Here we describe the crystal structure of a human CYP450, CYP2C9, both unliganded and in complex with the anti-coagulant drug warfarin. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 12861225-7 2003 The structure defines unanticipated interactions between CYP2C9 and warfarin, and reveals a new binding pocket. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 12861225-8 2003 The binding mode of warfarin suggests that CYP2C9 may undergo an allosteric mechanism during its function. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 12814975-0 2003 A new class of CYP2C9 inhibitors: probing 2C9 specificity with high-affinity benzbromarone derivatives. Benzbromarone 77-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 12844133-1 2003 AIM: Our goal was to investigate whether artemisinin autoinduction is caused by an increase in cytochrome P450 (CYP) 2B6 or CYP2C9 activities, we evaluated the effects of multiple-dose artemisinin administration on S-mephenytoin N-demethylation in healthy subjects. artemisinin 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 12844133-4 2003 The CYP2C9 in vivo marker tolbutamide was administered on day -28 and on days 7, 12, and 17 to monitor the minor involvement of CYP2C9 in S-mephenytoin N-demethylation. Tolbutamide 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12893985-5 2003 A more than two-fold reduced oral clearance in homozygous carriers of CYP2C9*3 was seen for celecoxib compared to carriers of the wild-type genotype CYP2C9*1/*1 and heterozygous carriers of one *3 allele were in-between (P = 0.003 for trend), whereas CYP2C9*2 had no significant influence on celecoxib pharmacokinetics. Celecoxib 92-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 12893985-5 2003 A more than two-fold reduced oral clearance in homozygous carriers of CYP2C9*3 was seen for celecoxib compared to carriers of the wild-type genotype CYP2C9*1/*1 and heterozygous carriers of one *3 allele were in-between (P = 0.003 for trend), whereas CYP2C9*2 had no significant influence on celecoxib pharmacokinetics. Celecoxib 292-301 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 12893985-6 2003 Decreased concentrations of carboxy- and hydroxy-celecoxib in heterozygous and homozygous carriers of CYP2C9*3 were detected which supported the influence of CYP2C9 polymorphisms on celecoxib pharmacokinetic variability. carboxy- and hydroxy-celecoxib 28-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 12893985-6 2003 Decreased concentrations of carboxy- and hydroxy-celecoxib in heterozygous and homozygous carriers of CYP2C9*3 were detected which supported the influence of CYP2C9 polymorphisms on celecoxib pharmacokinetic variability. carboxy- and hydroxy-celecoxib 28-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 12893985-6 2003 Decreased concentrations of carboxy- and hydroxy-celecoxib in heterozygous and homozygous carriers of CYP2C9*3 were detected which supported the influence of CYP2C9 polymorphisms on celecoxib pharmacokinetic variability. Celecoxib 49-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 12893985-6 2003 Decreased concentrations of carboxy- and hydroxy-celecoxib in heterozygous and homozygous carriers of CYP2C9*3 were detected which supported the influence of CYP2C9 polymorphisms on celecoxib pharmacokinetic variability. Celecoxib 49-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 12893985-7 2003 Approximately 0.5% of Caucasians carrying the genotype CYP2C9*3/*3 will have greatly increased internal exposure to celecoxib. Celecoxib 116-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 12814975-2 2003 The CYP2C9 pharmacophore is proposed to include either an anionic group or hydrogen bond donor in addition to its hydrophobic groups. Hydrogen 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12734606-1 2003 OBJECTIVE: This study analyzed the frequency of CYP2C9 variant alleles and evaluated the impact of CYP2C9 genotype on diclofenac metabolism in a Spanish population. Diclofenac 118-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12844136-0 2003 Acenocoumarol pharmacokinetics in relation to cytochrome P450 2C9 genotype. Acenocoumarol 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 12844136-1 2003 BACKGROUND AND OBJECTIVES: Cytochrome P450 (CYP) 2C9 is one of the major CYP enzymes involved in the biotransformation of drugs, among others, the oral anticoagulant acenocoumarol. Acenocoumarol 166-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-52 12844136-3 2003 Patients with the CYP2C9*3 variant are known to require a lower maintenance dose of racemic acenocoumarol. Acenocoumarol 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 12844136-4 2003 We investigated the impact of the polymorphisms CYP2C9*2 and CYP2C9*3 on the pharmacokinetics of R- and S-acenocoumarol. r- and s-acenocoumarol 97-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 12844136-4 2003 We investigated the impact of the polymorphisms CYP2C9*2 and CYP2C9*3 on the pharmacokinetics of R- and S-acenocoumarol. r- and s-acenocoumarol 97-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12844136-9 2003 The mean oral clearance of S-acenocoumarol was 45% lower in the CYP2C9*1/*3 genotypes (10.9 +/- 3.0 L/h versus 19.8 +/- 3.1 L/h, P =.02). Acenocoumarol 27-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 12844136-16 2003 CONCLUSION: The results show S-acenocoumarol pharmacokinetics to be dependent on CYP2C9 polymorphism. Acenocoumarol 29-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 12844136-17 2003 In particular, the presence of the CYP2C9*3 allele impairs oral clearance of the coumarin. coumarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 14527094-1 2003 Diclofenac has been used for the evaluation of CYP2C9 activity in vitro as well as in vivo with varying results. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). 4"-oh diclofenac 32-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). 4"-oh diclofenac 32-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). 4"-oh diclofenac 32-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-5 2003 The mean (+/- SD) of diclofenac/4"-OH diclofenac ratio was 1.5 times higher among subjects carrying CYP2C9*3 allele (CYP2C9*1/*3 and CYP2C9*2/*3 genotypes) (0.91 +/- 0.28), compared to CYP2C9*1/*1 subjects (0.60 +/- 0.11). 4"-oh diclofenac 32-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 14527094-6 2003 The results show that the urinary diclofenac/4"-OH diclofenac ratio might be used to study CYP2C9 in humans. Diclofenac 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 14527094-6 2003 The results show that the urinary diclofenac/4"-OH diclofenac ratio might be used to study CYP2C9 in humans. 4"-oh diclofenac 45-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12742136-0 2003 Analysis of diclofenac and its metabolites by high-performance liquid chromatography: relevance of CYP2C9 genotypes in diclofenac urinary metabolic ratios. Diclofenac 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12742136-2 2003 The polymorphic CYP2C9 is involved in the metabolism of diclofenac to 4"-OH diclofenac and 3"-OH diclofenac. Diclofenac 56-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 12742136-2 2003 The polymorphic CYP2C9 is involved in the metabolism of diclofenac to 4"-OH diclofenac and 3"-OH diclofenac. 4"-oh diclofenac 70-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 12742136-2 2003 The polymorphic CYP2C9 is involved in the metabolism of diclofenac to 4"-OH diclofenac and 3"-OH diclofenac. 3"-oh diclofenac 91-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 12742136-7 2003 The relationship between diclofenac metabolic ratios among different CYP2C9 genotypes is reported. Diclofenac 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 12742136-8 2003 The CYP2C9*3/*3 subject had the highest diclofenac/4"-OH ratios. Diclofenac 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12742136-8 2003 The CYP2C9*3/*3 subject had the highest diclofenac/4"-OH ratios. 4"-oh 51-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12728288-15 2003 CONCLUSIONS: CYP2C9*3 variants are associated with an increased bleeding risk in patients anticoagulated with phenprocoumon. Phenprocoumon 110-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12734606-6 2003 The diclofenac/4"-OH diclofenac urinary ratio, but not the diclofenac/3"-OH diclofenac and diclofenac/5-OH diclofenac ratios, was related to CYP2C9 genotype. Diclofenac 4-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 12734606-6 2003 The diclofenac/4"-OH diclofenac urinary ratio, but not the diclofenac/3"-OH diclofenac and diclofenac/5-OH diclofenac ratios, was related to CYP2C9 genotype. 4"-oh diclofenac 15-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 12734606-6 2003 The diclofenac/4"-OH diclofenac urinary ratio, but not the diclofenac/3"-OH diclofenac and diclofenac/5-OH diclofenac ratios, was related to CYP2C9 genotype. Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 12734606-6 2003 The diclofenac/4"-OH diclofenac urinary ratio, but not the diclofenac/3"-OH diclofenac and diclofenac/5-OH diclofenac ratios, was related to CYP2C9 genotype. Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 12734606-9 2003 CYP2C9*3 allele seems to influence the 4"-hydroxylation of diclofenac, although there is a large overlapping in the urinary metabolic ratio between the genotype groups studied Diclofenac 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 12695341-10 2003 These data indicate that voriconazole is a substrate for CYP2C9, CYP2C19, and CYP3A4, with CYP2C9 involvement being minimal in human liver microsomes. Voriconazole 25-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 14739630-3 2003 Genetic variants associated with the metabolism of (S)-warfarin by cytochrome P450 2C9 may have specific implications on untoward effects. Warfarin 51-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-86 14739630-5 2003 Studies have demonstrated that initial dosing of warfarin with CYP2C9*3 with a five-milligram dose caused an increase in the international normalized ratio and significant risk of bleeding. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 12800253-0 2003 Rapid detection of the known SNPs of CYP2C9 using oligonucleotide microarray. Oligonucleotides 50-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 12800253-3 2003 In order to facilitate the detection of the known SNPs of CYP2C9, an allele-specific oligonucleotide (ASO) based microarray was made. Oligonucleotides 85-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 12800253-3 2003 In order to facilitate the detection of the known SNPs of CYP2C9, an allele-specific oligonucleotide (ASO) based microarray was made. 1,5-anhydroglucitol 102-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 12800253-4 2003 METHODS: An oligonucleotide microarray was made to facilitate the SNP (single nucleotide polymorphism) screening and was applied for the detection of CYP2C9 polymorphism in 62 high blood pressure (HBP) patients who received Irbesartan for treatment. Oligonucleotides 12-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 12800253-6 2003 And the relation between CYP2C9 polymorphism and therapeutic outcome of Irbesartan was statistically analyzed. Irbesartan 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 12800253-11 2003 CONCLUSION: The oligonucleotide microarray made in this study is a reliable assay for detecting the CYP2C9 known alleles and the heterozygous CYP2C9*1/*3 has no significant effects on the therapeutic outcome of Irbesartan. Oligonucleotides 16-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 12800253-11 2003 CONCLUSION: The oligonucleotide microarray made in this study is a reliable assay for detecting the CYP2C9 known alleles and the heterozygous CYP2C9*1/*3 has no significant effects on the therapeutic outcome of Irbesartan. Irbesartan 211-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 12867215-12 2003 However, as 80% of voriconazole is hepatically eliminated, primarily via the cytochrome P450 (CYP) isozymes CYP2C19, CYP3A4, and CYP2C9, voriconazole has a high potential for drug interactions, and dose reduction is recommended in patients with mild to moderate hepatic dysfunction (Child-Pugh class A or B). Voriconazole 19-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 12684728-2 2003 The aim of this study was to investigate the contribution of CYP2C9 and CYP2C19 and their polymorphisms to the variability of cyclophosphamide activation. Cyclophosphamide 126-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12684728-4 2003 The kinetic data obtained in the yeast system revealed that the intrinsic clearance (V(max)/K(m)) of cyclophosphamide by CYP2C9.2 and CYP2C9.3 samples was approximately threefold lower than that by CYP2C9.1. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 12684728-4 2003 The kinetic data obtained in the yeast system revealed that the intrinsic clearance (V(max)/K(m)) of cyclophosphamide by CYP2C9.2 and CYP2C9.3 samples was approximately threefold lower than that by CYP2C9.1. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 12684728-4 2003 The kinetic data obtained in the yeast system revealed that the intrinsic clearance (V(max)/K(m)) of cyclophosphamide by CYP2C9.2 and CYP2C9.3 samples was approximately threefold lower than that by CYP2C9.1. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 12684728-7 2003 No correlation was found between 4-hydroxylation of cyclophosphamide and the formation rate of hydroxycelecoxib, mainly catalysed by CYP2C9 ( r(s)=0.17, P=0.55, n=32). Hydroxy Celecoxib 95-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 12820813-0 2003 Losartan and E3174 pharmacokinetics in cytochrome P450 2C9*1/*1, *1/*2, and *1/*3 individuals. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 12820813-0 2003 Losartan and E3174 pharmacokinetics in cytochrome P450 2C9*1/*1, *1/*2, and *1/*3 individuals. losartan carboxylic acid 13-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 12820813-8 2003 A significant association between CYP2C9 genotype and losartan to E3174 formation clearance was observed, such that 50% of the variability was accounted for by the genotype. Losartan 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 12820813-8 2003 A significant association between CYP2C9 genotype and losartan to E3174 formation clearance was observed, such that 50% of the variability was accounted for by the genotype. losartan carboxylic acid 66-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 12820813-11 2003 Alterations in losartan dosing in CYP2C9*1/*2 and *1/*3 individuals does not appear necessary. Losartan 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 12708941-12 2003 Phenytoin induces the metabolism of drugs that are substrates of CYP2C, CYP2D, and CYP3A; however, phenytoin is eliminated predominantly by CYP2C9- and CYP2C19-dependent hepatic metabolism. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 12708941-12 2003 Phenytoin induces the metabolism of drugs that are substrates of CYP2C, CYP2D, and CYP3A; however, phenytoin is eliminated predominantly by CYP2C9- and CYP2C19-dependent hepatic metabolism. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 12708941-16 2003 CONCLUSIONS: Phenytoin is a known inducer of drugs metabolized by CYP2C, CYP2D, and CYP3A, but its own metabolism may be altered by drugs influencing CYP2C9 or CYP2C19, such as diazepam. Phenytoin 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 12708941-16 2003 CONCLUSIONS: Phenytoin is a known inducer of drugs metabolized by CYP2C, CYP2D, and CYP3A, but its own metabolism may be altered by drugs influencing CYP2C9 or CYP2C19, such as diazepam. Diazepam 177-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 12695341-10 2003 These data indicate that voriconazole is a substrate for CYP2C9, CYP2C19, and CYP3A4, with CYP2C9 involvement being minimal in human liver microsomes. Voriconazole 25-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12724615-3 2003 The most important of these is CYP2C9, which hydroxylates the S-enantiomers of warfarin, acenocoumarol and phenprocoumon with high catalytic activity. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12682803-3 2003 The present study aimed to evaluate the influence of CYP2D6 and CYP2C9 genotypes, and tobacco smoking on steady-state thioridazine plasma levels. Thioridazine 118-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 12724615-3 2003 The most important of these is CYP2C9, which hydroxylates the S-enantiomers of warfarin, acenocoumarol and phenprocoumon with high catalytic activity. Acenocoumarol 89-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12724615-3 2003 The most important of these is CYP2C9, which hydroxylates the S-enantiomers of warfarin, acenocoumarol and phenprocoumon with high catalytic activity. Phenprocoumon 107-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12724615-4 2003 In at least eight separate clinical studies, possession of the CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, has been associated with a significant decrease in a mean warfarin dose requirement. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 12724615-4 2003 In at least eight separate clinical studies, possession of the CYP2C9*2 or CYP2C9*3 variant alleles, which result in decreased enzyme activity, has been associated with a significant decrease in a mean warfarin dose requirement. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 12724615-6 2003 Possession of the CYP2C9*3 variant also appears to be associated with a low acenocoumarol dose requirement. Acenocoumarol 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 12721102-22 2003 Of the other isoforms studied, CYP2C9 and CYP3A4 contribute to a lesser degree to promazine 5-sulphoxidation and N-demethylation, respectively. Promazine 82-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12698304-0 2003 Differences in flurbiprofen pharmacokinetics between CYP2C9*1/*1, *1/*2, and *1/*3 genotypes. Flurbiprofen 15-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 12642470-7 2003 Inhibition studies conducted for CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP2A6, and CYP2E1 showed troglitazone to be the most nonselective and potent inhibitor followed by rosiglitazone and pioglitazone. Troglitazone 108-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 12642470-9 2003 Troglitazone, in addition, inhibited CYP2C9 (K(i) 0.6 microM). Troglitazone 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. 4'-hydroxyflurbiprofen 252-274 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12698304-1 2003 OBJECTIVE: This study was conducted to examine differences in flurbiprofen metabolism among individuals with the CYP2C9*1/*1, *1/*2, and *1/*3 genotypes. Flurbiprofen 62-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. Flurbiprofen 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. 4'-hydroxyflurbiprofen 337-359 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12698304-5 2003 Flurbiprofen AUC(0- infinity )was significantly higher and all measures of flurbiprofen clearance were significantly lower in the CYP2C9*1/*3 individuals than in those with *1/*1. Flurbiprofen 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 12698304-5 2003 Flurbiprofen AUC(0- infinity )was significantly higher and all measures of flurbiprofen clearance were significantly lower in the CYP2C9*1/*3 individuals than in those with *1/*1. Flurbiprofen 75-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 12698304-7 2003 CONCLUSIONS: CYP2C9 genotype is a significant predictor of flurbiprofen disposition in humans by altering CYP2C9-mediated metabolism and reducing systemic clearance. Flurbiprofen 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. Flurbiprofen 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. Flurbiprofen 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12698304-4 2003 RESULTS: CYP2C9 genotype was a significant predictor of flurbiprofen metabolism and accounted for 59% of the variability in flurbiprofen AUC(0- infinity ), and approximately 50% of the variability in flurbiprofen oral clearance, formation clearance to 4"-hydroxyflurbiprofen, and the 0 to 24-h urinary metabolic ratio of flurbiprofen to 4"-hydroxyflurbiprofen. Flurbiprofen 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 12586744-4 2003 CYP 2C9 overexpression was associated with the activation of Akt and an increase in cyclin D1 expression, effects that were abolished by the epidermal growth factor (EGF) receptor inhibitor, AG1478, which also prevented the CYP 2C9-induced increase in cell proliferation. RTKI cpd 191-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-231 12698304-7 2003 CONCLUSIONS: CYP2C9 genotype is a significant predictor of flurbiprofen disposition in humans by altering CYP2C9-mediated metabolism and reducing systemic clearance. Flurbiprofen 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 12586744-0 2003 Cytochrome P450 2C9-derived epoxyeicosatrienoic acids induce angiogenesis via cross-talk with the epidermal growth factor receptor (EGFR). epoxyeicosatrienoic acids 28-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 12586744-5 2003 Stimulation of EGF receptor overexpressing cells with 11,12-EET or transfection of these cells with CYP 2C9 enhanced the tyrosine phosphorylation of the EGF receptor. Tyrosine 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-107 12586744-3 2003 In human endothelial cells overexpressing CYP 2C9, cell number was increased compared with control cells and was inhibited by the CYP 2C9 inhibitor, sulfaphenazole. Sulfaphenazole 149-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-49 12621390-0 2003 Population differences in S-warfarin metabolism between CYP2C9 genotype-matched Caucasian and Japanese patients. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 12586744-3 2003 In human endothelial cells overexpressing CYP 2C9, cell number was increased compared with control cells and was inhibited by the CYP 2C9 inhibitor, sulfaphenazole. Sulfaphenazole 149-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-137 12586744-4 2003 CYP 2C9 overexpression was associated with the activation of Akt and an increase in cyclin D1 expression, effects that were abolished by the epidermal growth factor (EGF) receptor inhibitor, AG1478, which also prevented the CYP 2C9-induced increase in cell proliferation. RTKI cpd 191-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 12649384-2 2003 Cytochromes P450 1A1 (18.5), 1A2 (20), and 2D6 (100) catalyzed the O-demethylation of harmaline, and CYP1A1 (98.5), CYP1A2 (35), CYP2C9 (16), CYP2C19 (30), and CYP2D6 (115) catalyzed that of harmine (relative activities). Harmaline 86-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 12649384-5 2003 K(cat) (min(-1)) and K(m) Awake M) values for harmaline were: CYP1A1, 10.8 and 11.8; CYP1A2, 12.3 and 13.3; CYP2C9, 5.3 and 175; CYP2C19, 10.3 and 160; and CYP2D6, 39.9 and 1.4. Harmaline 46-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 12649384-6 2003 Values for harmine were: CYP1A1, 45.2 and 52.2; CYP1A2, 9.2 and 14.7; CYP2C9, 11.9 and 117; CYP2C19, 21.4 and 121; and CYP2D6, 29.7 and 7.4. Harmine 11-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 12734935-9 2003 The time-dependent studies showed that PL had the stereoselectivity of S-(-)-isomer in metabolism via CYP2C18 and the stereoselectivity of R-(+)-isomer in metabolism via CYP2C9. Propranolol 39-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 12734935-9 2003 The time-dependent studies showed that PL had the stereoselectivity of S-(-)-isomer in metabolism via CYP2C18 and the stereoselectivity of R-(+)-isomer in metabolism via CYP2C9. BINOL, naphthol 139-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 12482324-1 2003 Human cytochrome P450 2C9 (CYP2C9) is important in the metabolism of non-steroidal anti-inflammatory compounds such as diclofenac, the antidiabetic agent tolbutamide and other clinically important drugs, many of which are weakly acidic. Diclofenac 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 12482324-1 2003 Human cytochrome P450 2C9 (CYP2C9) is important in the metabolism of non-steroidal anti-inflammatory compounds such as diclofenac, the antidiabetic agent tolbutamide and other clinically important drugs, many of which are weakly acidic. Diclofenac 119-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 12482324-1 2003 Human cytochrome P450 2C9 (CYP2C9) is important in the metabolism of non-steroidal anti-inflammatory compounds such as diclofenac, the antidiabetic agent tolbutamide and other clinically important drugs, many of which are weakly acidic. Tolbutamide 154-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-25 12482324-1 2003 Human cytochrome P450 2C9 (CYP2C9) is important in the metabolism of non-steroidal anti-inflammatory compounds such as diclofenac, the antidiabetic agent tolbutamide and other clinically important drugs, many of which are weakly acidic. Tolbutamide 154-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 12482324-2 2003 Multiple sequence alignment of CYPs identified CYP2C9 Asp(293) as corresponding to Asp(301) of CYP2D6, which has been suggested to play a role in the binding of basic substrates to the latter enzyme. Aspartic Acid 54-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12482324-2 2003 Multiple sequence alignment of CYPs identified CYP2C9 Asp(293) as corresponding to Asp(301) of CYP2D6, which has been suggested to play a role in the binding of basic substrates to the latter enzyme. Aspartic Acid 83-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12482324-8 2003 Our analysis suggests a structural role for the carboxyl side chain of Asp(293) in CYP2C9 substrate binding and catalysis. Aspartic Acid 71-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 12615570-4 2003 DISCUSSION: adverse outcomes with warfarin therapy could be explained and possibly avoided by identifying patients with variant alleles for CYP2C9 before initiation of therapy. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 12621390-1 2003 OBJECTIVE: Our objective was to investigate population differences in the metabolic activity of cytochrome P450 (CYP) 2C9 between genotypically matched Caucasian and Japanese patients by using the unbound oral clearance of S-warfarin as an in vivo phenotypic trait measure. Sulfur 223-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-121 12621390-1 2003 OBJECTIVE: Our objective was to investigate population differences in the metabolic activity of cytochrome P450 (CYP) 2C9 between genotypically matched Caucasian and Japanese patients by using the unbound oral clearance of S-warfarin as an in vivo phenotypic trait measure. Warfarin 225-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-121 12621390-9 2003 In addition, Japanese patients heterozygous for the CYP2C9*3 genotype (n = 4) showed a significantly (P <.05) reduced unbound oral clearance for S-warfarin, by 63%, as compared with Japanese patients possessing the homozygous CYP2C9*1 genotype. Warfarin 150-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 12637241-18 2003 Correlation analysis suggested that CYP2B11 catalyses the N-demethylation of dextromethorphan (mediated in humans by CYP3A) and the 4"-hydroxylation of mephenytoin (mediated in humans by CYP2C19) in the dog, and that this enzyme and CYP3A12 contribute to S-warfarin 7-hydroxylation (mediated in humans by CYP2C9). Nitrogen 58-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 305-311 12639175-8 2003 Drug interactions occur through inhibition of the CYP2C9, CYP2C19, and CYP3A4 isoenzymes, resulting in alterations in kinetic parameters of either voriconazole or the interacting agent. Voriconazole 147-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 12713683-7 2003 In addition, bosentan is not only a substrate but also an inducer of CYP3A4 and CYP2C9. Bosentan 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 12637241-5 2003 The rate of 7-hydroxylation of S-warfarin (CYP2C9 in humans) by dog liver microsomes (mean +/- SD from 12 (six male and six female) dogs = 10.8 +/- 1.9 fmol mg(-1) protein min(-1)) was 1.5-2 orders of magnitude lower than that in humans. Warfarin 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 12573493-2 2003 There is strong evidence suggesting that the activation of a CYP 2C epoxygenase in endothelial cells is an essential step in nitric oxide (NO)- and prostacyclin (PGI(2))-independent vasodilatation of several vascular beds, particularly in the heart and kidney. Nitric Oxide 125-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12573493-2 2003 There is strong evidence suggesting that the activation of a CYP 2C epoxygenase in endothelial cells is an essential step in nitric oxide (NO)- and prostacyclin (PGI(2))-independent vasodilatation of several vascular beds, particularly in the heart and kidney. Epoprostenol 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12573493-2 2003 There is strong evidence suggesting that the activation of a CYP 2C epoxygenase in endothelial cells is an essential step in nitric oxide (NO)- and prostacyclin (PGI(2))-independent vasodilatation of several vascular beds, particularly in the heart and kidney. Epoprostenol 162-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12573493-6 2003 Cerivastatin and fluvastatin increase CYP 2C mRNA and protein in native and cultured endothelial cells, and enhance the bradykinin-induced NO/PGI(2)-independent relaxation of arterial segments as well as the generation of reactive oxygen species. cerivastatin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 12573493-6 2003 Cerivastatin and fluvastatin increase CYP 2C mRNA and protein in native and cultured endothelial cells, and enhance the bradykinin-induced NO/PGI(2)-independent relaxation of arterial segments as well as the generation of reactive oxygen species. Fluvastatin 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 12446689-9 2003 Intriguingly, removal of the negative charge from either 216 or 301 produced enzymes (E216A, E216K, and D301Q) with elevated levels (50-75-fold) of catalytic activity toward diclofenac, a carboxylate-containing CYP2C9 substrate that lacks a basic nitrogen atom. Diclofenac 174-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 12446689-9 2003 Intriguingly, removal of the negative charge from either 216 or 301 produced enzymes (E216A, E216K, and D301Q) with elevated levels (50-75-fold) of catalytic activity toward diclofenac, a carboxylate-containing CYP2C9 substrate that lacks a basic nitrogen atom. Nitrogen 247-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 12446689-11 2003 The kinetic parameters for diclofenac (K(m) 108 microm, k(cat) 5 min(-1)) along with nifedipine (K(m) 28 microm, k(cat) 2 min(-1)) and tolbutamide (K(m) 315 microm, k(cat) 1 min(-1)), which are not normally substrates for CYP2D6, were within an order of magnitude of those observed with CYP3A4 or CYP2C9. Diclofenac 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 297-303 12549950-10 2003 Fenofibrate is also a mild to moderate inhibitor of CYP2C9, the enzyme responsible for warfarin metabolism. Fenofibrate 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 12549950-10 2003 Fenofibrate is also a mild to moderate inhibitor of CYP2C9, the enzyme responsible for warfarin metabolism. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 12559973-1 2003 Studies were designed to investigate the possible mechanisms associated with the kinetic observation of CYP2C9 activation by dapsone and its phase I metabolite, N-hydroxydapsone. Dapsone 125-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 12559973-1 2003 Studies were designed to investigate the possible mechanisms associated with the kinetic observation of CYP2C9 activation by dapsone and its phase I metabolite, N-hydroxydapsone. 4-amino-4'-hydroxylaminodiphenylsulfone 161-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 12559973-2 2003 Kinetic studies suggested that dapsone activated CYP2C9-mediated flurbiprofen 4(")-hydroxylation by decreasing the K(m) (alpha=0.2) and increasing the V(max) (beta=1.9). Dapsone 31-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 12559973-2 2003 Kinetic studies suggested that dapsone activated CYP2C9-mediated flurbiprofen 4(")-hydroxylation by decreasing the K(m) (alpha=0.2) and increasing the V(max) (beta=1.9). Flurbiprofen 65-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 12559973-5 2003 In the presence of dapsone, the spectral binding constant (K(s)) for flurbiprofen was reduced from 14.1 to 2.1 microM, while in the presence of N-hydroxydapsone, the K(s) remained unchanged (14.0 microM), which suggests that dapsone causes an increase in the affinity of flurbiprofen for CYP2C9, whereas N-hydroxydapsone does not. Dapsone 19-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 288-294 12559973-5 2003 In the presence of dapsone, the spectral binding constant (K(s)) for flurbiprofen was reduced from 14.1 to 2.1 microM, while in the presence of N-hydroxydapsone, the K(s) remained unchanged (14.0 microM), which suggests that dapsone causes an increase in the affinity of flurbiprofen for CYP2C9, whereas N-hydroxydapsone does not. Flurbiprofen 69-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 288-294 12559973-7 2003 Interestingly, the presence of N-hydroxydapsone generally caused the same effects on stoichiometry as those of flurbiprofen 4(")-hydroxylation but failed to reduce excess water formation, which suggests that, while N-hydroxydapsone activates CYP2C9, it does so less efficiently and possibly through a mechanism different from that of dapsone. Dapsone 40-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 242-248 12450521-8 2003 This result suggests the implication of genetically polymorphic enzymes, presumably CYP2D6, CYP2C9 and CYP2C19 in the metabolism of fluoxetine to norfluoxetine. Fluoxetine 132-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 12450521-8 2003 This result suggests the implication of genetically polymorphic enzymes, presumably CYP2D6, CYP2C9 and CYP2C19 in the metabolism of fluoxetine to norfluoxetine. norfluoxetine 146-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 12388259-5 2003 Finally, we tested the most abundant arachidonic acid metabolite of CYP2C9, 14,15-epoxyeicosatrienoic acid, which induced angiogenesis in vivo when embedded in Matrigel plugs and implanted in adult rats. Arachidonic Acid 37-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 12388259-5 2003 Finally, we tested the most abundant arachidonic acid metabolite of CYP2C9, 14,15-epoxyeicosatrienoic acid, which induced angiogenesis in vivo when embedded in Matrigel plugs and implanted in adult rats. 14,15-epoxy-5,8,11-eicosatrienoic acid 76-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 12464242-2 2003 From a consideration of specific interactions between drug substrates for human CYP2 family enzymes and the putative active sites of CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP2E1, it is likely that the number and disposition of hydrogen bond donor/acceptors and aromatic rings within the various P450 substrate molecules determines their enzyme selectivity and binding affinity, together with directing their preferred routes of metabolism by the CYP2 enzymes concerned. Hydrogen 242-250 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 12464247-0 2003 Substrate selectivity of human cytochrome P450 2C9: importance of residues 476, 365, and 114 in recognition of diclofenac and sulfaphenazole and in mechanism-based inactivation by tienilic acid. Diclofenac 111-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-50 12464247-0 2003 Substrate selectivity of human cytochrome P450 2C9: importance of residues 476, 365, and 114 in recognition of diclofenac and sulfaphenazole and in mechanism-based inactivation by tienilic acid. Sulfaphenazole 126-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-50 12464247-0 2003 Substrate selectivity of human cytochrome P450 2C9: importance of residues 476, 365, and 114 in recognition of diclofenac and sulfaphenazole and in mechanism-based inactivation by tienilic acid. Ticrynafen 180-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-50 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Ticrynafen 294-307 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Ticrynafen 294-307 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-181 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Ticrynafen 309-311 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Ticrynafen 309-311 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-181 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Diclofenac 379-389 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Diclofenac 379-389 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-181 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Sulfaphenazole 449-463 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-48 12464247-1 2003 A series of six site-directed mutants of CYP 2C9 were constructed with the aim to better define the amino acid residues that play a critical role in substrate selectivity of CYP 2C9, particularly in three distinctive properties of this enzyme: (i) its selective mechanism-based inactivation by tienilic acid (TA), (ii) its high affinity and hydroxylation regioselectivity toward diclofenac, and (iii) its high affinity for the competitive inhibitor sulfaphenazole (SPA). Sulfaphenazole 449-463 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-181 12464247-5 2003 At the opposite end, the F94L and F110I mutants showed properties very similar to those of CYP 2C9 toward TA and diclofenac. Ticrynafen 106-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 12464247-5 2003 At the opposite end, the F94L and F110I mutants showed properties very similar to those of CYP 2C9 toward TA and diclofenac. Diclofenac 113-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 12464247-7 2003 These data indicate that Phe 114 plays an important role in recognition of aromatic substrates of CYP 2C9, presumably via Pi-stacking interactions. Phenylalanine 25-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-105 12464247-8 2003 They also provide the first experimental evidence showing that Phe 476 plays a crucial role in substrate recognition and hydroxylation by CYP 2C9. Phenylalanine 63-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-145 14558433-8 2003 Likewise, fluconazole interferes with the CYP2C9-mediated hepatic elimination of fluvastatin. Fluconazole 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 14558433-8 2003 Likewise, fluconazole interferes with the CYP2C9-mediated hepatic elimination of fluvastatin. Fluvastatin 81-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 14650362-6 2003 The cytochrome P (CYP) enzyme CYP2C9 plays a major role in the metabolism of gemfibrozil. Gemfibrozil 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 12634980-1 2003 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) allelic variant carriers have been shown to experience hyper-responsiveness to small doses of oral anticoagulants (OAs) (warfarin or acenocoumarol) and a higher bleeding rate. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-31 12634980-1 2003 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) allelic variant carriers have been shown to experience hyper-responsiveness to small doses of oral anticoagulants (OAs) (warfarin or acenocoumarol) and a higher bleeding rate. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12634980-1 2003 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) allelic variant carriers have been shown to experience hyper-responsiveness to small doses of oral anticoagulants (OAs) (warfarin or acenocoumarol) and a higher bleeding rate. Acenocoumarol 174-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-31 12634980-1 2003 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) allelic variant carriers have been shown to experience hyper-responsiveness to small doses of oral anticoagulants (OAs) (warfarin or acenocoumarol) and a higher bleeding rate. Acenocoumarol 174-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12637241-18 2003 Correlation analysis suggested that CYP2B11 catalyses the N-demethylation of dextromethorphan (mediated in humans by CYP3A) and the 4"-hydroxylation of mephenytoin (mediated in humans by CYP2C19) in the dog, and that this enzyme and CYP3A12 contribute to S-warfarin 7-hydroxylation (mediated in humans by CYP2C9). Dextromethorphan 77-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 305-311 12637241-18 2003 Correlation analysis suggested that CYP2B11 catalyses the N-demethylation of dextromethorphan (mediated in humans by CYP3A) and the 4"-hydroxylation of mephenytoin (mediated in humans by CYP2C19) in the dog, and that this enzyme and CYP3A12 contribute to S-warfarin 7-hydroxylation (mediated in humans by CYP2C9). Mephenytoin 152-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 305-311 12637241-18 2003 Correlation analysis suggested that CYP2B11 catalyses the N-demethylation of dextromethorphan (mediated in humans by CYP3A) and the 4"-hydroxylation of mephenytoin (mediated in humans by CYP2C19) in the dog, and that this enzyme and CYP3A12 contribute to S-warfarin 7-hydroxylation (mediated in humans by CYP2C9). Warfarin 257-265 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 305-311 12603175-0 2003 Influence of age and cytochrome P450 2C9 genotype on the steady-state disposition of diclofenac and celecoxib. Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 12485954-5 2003 The metabolism of NE-100 was mediated with recombinant CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. N,N-dipropyl-2-(4-methoxy-3-(2-phenylethoxy)phenyl)ethylamine monohydrochloride 18-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 12534640-1 2003 AIMS: The cytochrome P450 enzyme CYP2C9 catalyses the 4"-hydroxylation of the nonsteroidal analgesic drug diclofenac in humans. Diclofenac 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12534640-2 2003 We studied the influences of the known amino acid variants, CYP2C9*2 (Arg144Cys) and CYP2C9*3 (Ile359Leu), on diclofenac pharmacokinetics after a 50-mg oral dose of diclofenac in healthy volunteers. Diclofenac 110-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 12534640-2 2003 We studied the influences of the known amino acid variants, CYP2C9*2 (Arg144Cys) and CYP2C9*3 (Ile359Leu), on diclofenac pharmacokinetics after a 50-mg oral dose of diclofenac in healthy volunteers. Diclofenac 110-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 12882588-10 2003 Rifampicin also induces CYP2C-mediated metabolism and thus reduces the plasma concentrations of, for example, the CYP2C9 substrate (S)-warfarin and the sulfonylurea antidiabetic drugs. Rifampin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 12882588-10 2003 Rifampicin also induces CYP2C-mediated metabolism and thus reduces the plasma concentrations of, for example, the CYP2C9 substrate (S)-warfarin and the sulfonylurea antidiabetic drugs. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 14768975-8 2003 This may be due to increased metabolism of celecoxib due to the induction of cytochrome P4502C9 (CYP2C9) in liver. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-95 14768975-8 2003 This may be due to increased metabolism of celecoxib due to the induction of cytochrome P4502C9 (CYP2C9) in liver. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 15618748-8 2003 ABT exhibited the highest Ki value for CYP2C9-dependent diclofenac 4"-hydroxylation among those determined by this assay (Ki=3500 microM). diclofenac 4" 56-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 12603175-0 2003 Influence of age and cytochrome P450 2C9 genotype on the steady-state disposition of diclofenac and celecoxib. Celecoxib 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 12603176-1 2003 BACKGROUND: In vitro, bosentan has been shown to be a mild inducer of cytochrome P450 (CYP) 2C9 and 3A4. Bosentan 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-95 14586772-4 2003 Aspirin-induced asthma (AIA) is a typical drug-induced phenotype due to aspirin or nonsteroidal antiinflammatory drugs, and these drugs are metabolized by CYP2C9 and UGT1A6, which are regulated by PXR. Aspirin 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 12558459-7 2003 Cyclosporin-treated patients on the other hand show several-fold higher systemic exposure of all statins, both those that are metabolised by CYP3A4 and fluvastatin (metabolised by CYP2C9). Cyclosporine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 12558459-7 2003 Cyclosporin-treated patients on the other hand show several-fold higher systemic exposure of all statins, both those that are metabolised by CYP3A4 and fluvastatin (metabolised by CYP2C9). Fluvastatin 152-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 12520632-0 2003 Tolbutamide, flurbiprofen, and losartan as probes of CYP2C9 activity in humans. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 12520632-0 2003 Tolbutamide, flurbiprofen, and losartan as probes of CYP2C9 activity in humans. Losartan 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 12520632-5 2003 The formation clearance of tolbutamide to its CYP2C9-mediated metabolites demonstrated a stronger association with genotype compared to flurbiprofen and losartan, respectively (r2 = 0.64 vs. 0.53 vs. 0.42). Tolbutamide 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 12520632-5 2003 The formation clearance of tolbutamide to its CYP2C9-mediated metabolites demonstrated a stronger association with genotype compared to flurbiprofen and losartan, respectively (r2 = 0.64 vs. 0.53 vs. 0.42). Losartan 153-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 12520632-8 2003 Tolbutamide is a better CYP2C9 probe than flurbiprofen and losartan, and the 0- to 12-hour amount of 4"-hydroxytolbutamide and carboxytolbutamide is the best urinary measure of its metabolism. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 14682608-5 2003 In the presence of gemfibrozil, a focal point was obtained in Dixon plots, demonstrating that there was inhibition of CYP2C8-, CYP2C9- and CYP3A4-mediated metabolism. Gemfibrozil 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 12509498-0 2003 Cytochrome P450 2C9 plays an important role in the regulation of exercise-induced skeletal muscle blood flow and oxygen uptake in humans. Oxygen 113-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 12509498-9 2003 The results demonstrate that CYP 2C9 plays an important role in the regulation of hyperaemia and oxygen uptake during exercise. Oxygen 97-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-36 12426520-6 2002 RESULTS: Tolbutamide area under the plasma concentration-time curve from time zero to infinity [AUC(0- infinity )] significantly increased by 1.5-fold and 1.9-fold, respectively, in subjects expressing the CYP2C9(*)1/(*)2 and (*)1/(*)3 genotypes compared with (*)1/(*)1 subjects. Tolbutamide 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 14583800-1 2003 Alteration of monoaminergic neurotransmission has been implicated in the pathophysiology of mood disorders, and CYP2C9 enzyme activity has been shown to be modulated by serotonin in vitro. Serotonin 169-178 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 14597963-0 2003 Influence of CYP2C9 genotypes on the formation of a hepatotoxic metabolite of valproic acid in human liver microsomes. Valproic Acid 78-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 14597963-1 2003 The present study investigated the effect of cytochrome P450 2C9 (CYP2C9) genetic polymorphism on the biotransformation of valproic acid (VPA) to its hepatotoxic metabolite, 4-ene-VPA, and compared that to the formation of the inactive 4-OH-VPA and 5-OH-VPA. Valproic Acid 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-64 14597963-1 2003 The present study investigated the effect of cytochrome P450 2C9 (CYP2C9) genetic polymorphism on the biotransformation of valproic acid (VPA) to its hepatotoxic metabolite, 4-ene-VPA, and compared that to the formation of the inactive 4-OH-VPA and 5-OH-VPA. Valproic Acid 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 14597963-1 2003 The present study investigated the effect of cytochrome P450 2C9 (CYP2C9) genetic polymorphism on the biotransformation of valproic acid (VPA) to its hepatotoxic metabolite, 4-ene-VPA, and compared that to the formation of the inactive 4-OH-VPA and 5-OH-VPA. Valproic Acid 138-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-64 14597963-1 2003 The present study investigated the effect of cytochrome P450 2C9 (CYP2C9) genetic polymorphism on the biotransformation of valproic acid (VPA) to its hepatotoxic metabolite, 4-ene-VPA, and compared that to the formation of the inactive 4-OH-VPA and 5-OH-VPA. Valproic Acid 138-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 2-propyl-4-pentenoic acid 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 2-propyl-4-pentenoic acid 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 239-245 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 4-Hydroxyvalproic acid 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 4-Hydroxyvalproic acid 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 239-245 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 5-Hydroxyvalproic acid 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 14597963-4 2003 The formation rates of 4-ene-VPA, 4-OH-VPA, and 5-OH-VPA in human liver microsomes were reduced by 29, 28, and 31%, respectively, in samples with one mutated CYP2C9 allele, and by 61, 73, and 58%, respectively, in samples with two mutated CYP2C9 alleles. 5-Hydroxyvalproic acid 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 239-245 12433806-8 2002 In addition, the selectivity of (-)-OSU6162 to inhibit six human p450 enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2E1, CYP2D6 and CYP3A4) was evaluated using an in vitro inhibition screen. OSU 6162 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 12386124-0 2002 Activation of CYP2C9-mediated metabolism by a series of dapsone analogs: kinetics and structural requirements. Dapsone 56-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 12386124-1 2002 Cytochrome P450 2C9-mediated metabolism has been shown to be activated in the presence of the effector dapsone. Dapsone 103-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 12386124-9 2002 Overall, it is apparent from these studies that a sulfone group in direct association with two benzene rings with para-electron-donating groups represents the most efficient activator of CYP2C9. Sulfones 50-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 12386124-9 2002 Overall, it is apparent from these studies that a sulfone group in direct association with two benzene rings with para-electron-donating groups represents the most efficient activator of CYP2C9. Benzene 95-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 12386132-2 2002 Phenytoin is oxidized to 4"-HPPH by CYP2C9 and to a minor extent by CYP2C19, and then 4"-HPPH is metabolized to 4"-HPPH O-glucuronide by UDP-glucuronosyltransferase (UGT). Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12386132-2 2002 Phenytoin is oxidized to 4"-HPPH by CYP2C9 and to a minor extent by CYP2C19, and then 4"-HPPH is metabolized to 4"-HPPH O-glucuronide by UDP-glucuronosyltransferase (UGT). hydroxyphenytoin 25-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12451430-1 2002 OBJECTIVE: To examine the effect of fluconazole, a potent inhibitor of CYP2C9 and CYP2C19, on the pharmacokinetics of rosuvastatin in healthy volunteers. Fluconazole 36-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 12451430-1 2002 OBJECTIVE: To examine the effect of fluconazole, a potent inhibitor of CYP2C9 and CYP2C19, on the pharmacokinetics of rosuvastatin in healthy volunteers. Rosuvastatin Calcium 118-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 12451434-3 2002 The lower activity of CYP2C9*5 encoded enzyme than *1 has been reported for the S-warfarin 7-hydroxylation in vitro. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 12822205-5 2003 Fluvastatin has a low potential for drug interactions due to its CYP2C9-dependant metabolism. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 12452751-7 2002 DISCUSSION: The metabolism of warfarin involves several cytochrome P450 isoenzymes, including CYP1A2, CYP2C9, CYP2C19, and CYP3A4. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 12452751-8 2002 Fluvoxamine has the potential to inhibit CYP1A2, CYP2C9, CYP2C19, and CYP3A4 to a significant degree. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 12496751-0 2002 Influence of CYP2C9 and CYP2C19 genetic polymorphisms on warfarin maintenance dose and metabolic clearance. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 12496751-1 2002 OBJECTIVE: Our objective was to determine the influence of cytochrome P450 (CYP) 2C9 and CYP2C19 genetic polymorphisms on warfarin dose requirement and metabolic clearance. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-84 12496751-8 2002 The mean S-warfarin CL(free) values varied significantly among the CYP2C9 genotype groups (P <.0001), although most patients (72%) with no mutated alleles showed S-warfarin CL(free) values in the same range as those carrying mutated alleles (58-777 mL/min). warfarin cl 11-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 12496751-8 2002 The mean S-warfarin CL(free) values varied significantly among the CYP2C9 genotype groups (P <.0001), although most patients (72%) with no mutated alleles showed S-warfarin CL(free) values in the same range as those carrying mutated alleles (58-777 mL/min). s-warfarin cl 9-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 12496751-10 2002 CONCLUSION: CYP2C9 genetic polymorphisms markedly influence warfarin dose requirements and metabolic clearance of the S-warfarin enantiomer, although nongenetic factors may also contribute to their large interindividual variability. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 12496751-10 2002 CONCLUSION: CYP2C9 genetic polymorphisms markedly influence warfarin dose requirements and metabolic clearance of the S-warfarin enantiomer, although nongenetic factors may also contribute to their large interindividual variability. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 12913403-0 2002 Warfarin dose adjustments based on CYP2C9 genetic polymorphisms. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 12913403-3 2002 The purpose of this study was to further characterize the relationship between CYP2C9 genotype and phenotype and to develop a basis for guidelines to interpret CYP2C9 genotype for warfarin dosing. Warfarin 180-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 12913403-3 2002 The purpose of this study was to further characterize the relationship between CYP2C9 genotype and phenotype and to develop a basis for guidelines to interpret CYP2C9 genotype for warfarin dosing. Warfarin 180-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 12913403-9 2002 Both CYP2C9*2 and *3 alleles were associated with lower maintenance dosages, lower total and R-warfarin plasma concentrations, decreased oral clearance of S-warfarin, increased plasma S:R-warfarin ratio and extended S-warfarin elimination half-life. Warfarin 93-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 12913403-9 2002 Both CYP2C9*2 and *3 alleles were associated with lower maintenance dosages, lower total and R-warfarin plasma concentrations, decreased oral clearance of S-warfarin, increased plasma S:R-warfarin ratio and extended S-warfarin elimination half-life. Sulfur 155-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 12913403-9 2002 Both CYP2C9*2 and *3 alleles were associated with lower maintenance dosages, lower total and R-warfarin plasma concentrations, decreased oral clearance of S-warfarin, increased plasma S:R-warfarin ratio and extended S-warfarin elimination half-life. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 12913403-9 2002 Both CYP2C9*2 and *3 alleles were associated with lower maintenance dosages, lower total and R-warfarin plasma concentrations, decreased oral clearance of S-warfarin, increased plasma S:R-warfarin ratio and extended S-warfarin elimination half-life. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 12913403-9 2002 Both CYP2C9*2 and *3 alleles were associated with lower maintenance dosages, lower total and R-warfarin plasma concentrations, decreased oral clearance of S-warfarin, increased plasma S:R-warfarin ratio and extended S-warfarin elimination half-life. Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 12913403-12 2002 The warfarin dose required to maintain a consistent target INR between subjects differs as a function of S-warfarin clearance which is decreased by both CYP2C9*2 and or CYP2C9*3 variant alleles. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 12913403-12 2002 The warfarin dose required to maintain a consistent target INR between subjects differs as a function of S-warfarin clearance which is decreased by both CYP2C9*2 and or CYP2C9*3 variant alleles. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 12913403-12 2002 The warfarin dose required to maintain a consistent target INR between subjects differs as a function of S-warfarin clearance which is decreased by both CYP2C9*2 and or CYP2C9*3 variant alleles. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 12913403-12 2002 The warfarin dose required to maintain a consistent target INR between subjects differs as a function of S-warfarin clearance which is decreased by both CYP2C9*2 and or CYP2C9*3 variant alleles. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. inn 10-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12426514-3 2002 Rifampin (INN, rifampicin), a potent enzyme inducer of CYP-mediated metabolism (especially CYP2C9, CYP2C19, and CYP3A4), is known to markedly decrease plasma concentrations and effects of a number coadministered drugs. Rifampin 15-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 12426520-10 2002 With tolbutamide used as a CYP2C9 probe, CYP2C9 genotype was the major determinant of CYP2C9 phenotype (r(2) = 0.77). Tolbutamide 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 12426520-10 2002 With tolbutamide used as a CYP2C9 probe, CYP2C9 genotype was the major determinant of CYP2C9 phenotype (r(2) = 0.77). Tolbutamide 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 12426520-10 2002 With tolbutamide used as a CYP2C9 probe, CYP2C9 genotype was the major determinant of CYP2C9 phenotype (r(2) = 0.77). Tolbutamide 5-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 12392591-2 2002 The aims of the present in vitro investigation were 1) to compare the rate of celecoxib hydroxylation by different genetic variants of cytochrome P450 2C9 (CYP2C9), and 2) to identify the enzyme(s) involved in the formation of the major metabolite carboxycelecoxib. Celecoxib 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-154 12414349-6 2002 Out of 170 patients requiring a low-dose of acenocoumarol (<or= 2 mg/day), 27.1% carried the CYP2C9*3 allele, while among the patients requiring higher doses, 8.4% had CYP2C9*3 (OR=4.77, 95% CI = 2.40-9.48, p<0.001 vs. 2C9*1/*1 patients). Acenocoumarol 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 12414349-6 2002 Out of 170 patients requiring a low-dose of acenocoumarol (<or= 2 mg/day), 27.1% carried the CYP2C9*3 allele, while among the patients requiring higher doses, 8.4% had CYP2C9*3 (OR=4.77, 95% CI = 2.40-9.48, p<0.001 vs. 2C9*1/*1 patients). Acenocoumarol 44-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 12414349-7 2002 In the multivariate analysis, independent predictive variables for low-dose acenocoumarol requirements were age >70 years (OR=3.73, 95%CI=2.29-6.08, p<0.001), and the CYP2C9*3 allele (OR=4.75, 95%CI=2.36-9.55, p <0.001). Acenocoumarol 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 12414349-9 2002 INTERPRETATION AND CONCLUSIONS: CYP2C9*3 is related to lower acenocoumarol dose requirements, a higher frequency of over-anticoagulation at the initiation of therapy and an unstable anticoagulant response. Acenocoumarol 61-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 12414349-0 2002 Pharmacogenetics of acenocoumarol: cytochrome P450 CYP2C9 polymorphisms influence dose requirements and stability of anticoagulation. Acenocoumarol 20-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 12414349-2 2002 Variant alleles, CYP2C9*2 and CYP2C9*3, have been related to decreased enzymatic activity, but their clinical relevance in acenocoumarol metabolism has not been established. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 12414349-2 2002 Variant alleles, CYP2C9*2 and CYP2C9*3, have been related to decreased enzymatic activity, but their clinical relevance in acenocoumarol metabolism has not been established. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 12414349-3 2002 We investigated CYP2C9 polymorphisms in relation to acenocoumarol dose requirement, stability of anticoagulation and bleeding. Acenocoumarol 52-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 12414349-4 2002 DESIGN AND METHODS: CYP2C9 genotyping was performed in 325 acenocoumarol-treated patients (INR target between 2.0 and 3.0) and in an additional group of 84 patients with repeated bleeding. Acenocoumarol 59-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 12414349-5 2002 RESULTS: Patients with the wild-type CYP2C9*1/*1 genotype (n=169) required a higher maintenance dose of acenocoumarol (17.1 8.7 mg/week) than did patients with the CYP2C9*2 (14.6 6.4 mg/week, p<0.05, N=97) or the CYP2C9*3 allele (11.2 6.2 mg/week, p<0.001, n=59). Acenocoumarol 104-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 12392591-2 2002 The aims of the present in vitro investigation were 1) to compare the rate of celecoxib hydroxylation by different genetic variants of cytochrome P450 2C9 (CYP2C9), and 2) to identify the enzyme(s) involved in the formation of the major metabolite carboxycelecoxib. Celecoxib 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 12392591-2 2002 The aims of the present in vitro investigation were 1) to compare the rate of celecoxib hydroxylation by different genetic variants of cytochrome P450 2C9 (CYP2C9), and 2) to identify the enzyme(s) involved in the formation of the major metabolite carboxycelecoxib. carboxycelecoxib 248-264 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-154 12392591-2 2002 The aims of the present in vitro investigation were 1) to compare the rate of celecoxib hydroxylation by different genetic variants of cytochrome P450 2C9 (CYP2C9), and 2) to identify the enzyme(s) involved in the formation of the major metabolite carboxycelecoxib. carboxycelecoxib 248-264 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 12392591-7 2002 RESULTS: The intrinsic clearance of celecoxib hydroxylation was significantly lower for yeast-expressed CYP2C9.3 (0.14 ml min-1 nmol-1 enzyme) compared with CYP2C9.1 (0.44 ml min-1 nmol-1 enzyme). Celecoxib 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 12392591-7 2002 RESULTS: The intrinsic clearance of celecoxib hydroxylation was significantly lower for yeast-expressed CYP2C9.3 (0.14 ml min-1 nmol-1 enzyme) compared with CYP2C9.1 (0.44 ml min-1 nmol-1 enzyme). Celecoxib 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 12392591-8 2002 In human liver microsomes, a significant 2-fold decrease in the rate of hydroxycelecoxib formation was evident in CYP2C9*1/*3 samples compared with CYP2C9*1/*1 samples. Hydroxy Celecoxib 72-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 12392591-8 2002 In human liver microsomes, a significant 2-fold decrease in the rate of hydroxycelecoxib formation was evident in CYP2C9*1/*3 samples compared with CYP2C9*1/*1 samples. Hydroxy Celecoxib 72-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 12392591-9 2002 There was also a marked reduction (up to 5.3 times) of hydroxycelecoxib formation in a liver sample genotyped as CYP2C9*3/*3. Hydroxy Celecoxib 55-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12392591-11 2002 Inhibition experiments with sulphaphenazole (SPZ) or triacetyloleandomycin indicated that celecoxib hydroxylation in human liver microsomes was mainly dependent on CYP2C9 and not CYP3A4. Celecoxib 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 12392591-14 2002 CONCLUSIONS: The results confirm that CYP2C9 is the major enzyme for celecoxib hydroxylation in vitro and further indicate that the CYP2C9*3 allelic variant is associated with markedly slower metabolism. Celecoxib 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 12392591-14 2002 CONCLUSIONS: The results confirm that CYP2C9 is the major enzyme for celecoxib hydroxylation in vitro and further indicate that the CYP2C9*3 allelic variant is associated with markedly slower metabolism. Celecoxib 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Irbesartan 206-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 12392591-0 2002 Oxidation of celecoxib by polymorphic cytochrome P450 2C9 and alcohol dehydrogenase. Celecoxib 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-57 12228186-5 2002 In the incubation study using cDNA-expressed P450s, both CYP2C9 and CYP2C19 catalyzed phenytoin p-hydroxylation, whereas TCAs inhibited only the CYP2C19 pathway. Phenytoin 86-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 12359989-0 2002 The CYP2C9 genotype predicts the blood pressure response to irbesartan: results from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation vs Atenolol (SILVHIA) trial. Irbesartan 60-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12359989-0 2002 The CYP2C9 genotype predicts the blood pressure response to irbesartan: results from the Swedish Irbesartan Left Ventricular Hypertrophy Investigation vs Atenolol (SILVHIA) trial. Atenolol 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Phenytoin 113-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Phenytoin 113-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Losartan 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Losartan 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12236850-0 2002 Involvement of CYP2C9 and UGT2B7 in the metabolism of zaltoprofen, a nonsteroidal anti-inflammatory drug, and its lack of clinically significant CYP inhibition potential. pyranoprofen 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 12236850-4 2002 The inhibitory effects of zaltoprofen on the metabolism of selective probe substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4 were also determined in human liver microsomes. pyranoprofen 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 12236850-5 2002 RESULTS: Zaltoprofen was extensively metabolized by CYP2C9 and UGT2B7. pyranoprofen 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 12236850-7 2002 In the human liver microsomal metabolism study, zaltoprofen metabolism was markedly inhibited by sulphaphenazole, a selective inhibitor of CYP2C9. pyranoprofen 48-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 12236850-7 2002 In the human liver microsomal metabolism study, zaltoprofen metabolism was markedly inhibited by sulphaphenazole, a selective inhibitor of CYP2C9. Sulfaphenazole 97-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 12236850-9 2002 However, zaltoprofen inhibited CYP2C9 by 26% at 5 micro g ml(-1). pyranoprofen 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 12236850-10 2002 At higher concentrations, zaltoprofen produced some inhibition of CYP2C9 (IC50 = 19.2 micro g ml(-1); 64.4 micro m) and CYP3A4 (IC50 = 53.9 micro g ml(-1); 181 micro m). pyranoprofen 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 12236850-12 2002 Furthermore, the maximum free drug concentrations in the hepatic intracellular was calculated to be 0.068 micro g ml(-1) and the increase in the AUC in the presence of zaltoprofen was estimated to be only 0.4% for CYP2C9 substrates and 0.1% for CYP3A4 substrates, respectively. pyranoprofen 168-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 214-220 12236850-13 2002 CONCLUSIONS: Zaltoprofen is predominantly metabolized by CYP2C9 and UGT2B7, and is considered unlikely to cause significant drug interactions in vivo when coadministered with CYP substrates at clinically effective doses. pyranoprofen 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 12231541-8 2002 ET743 was metabolized by cDNA-expressed human CYP3A4 and to a much lesser extent by CYP2C9, CYP2D6, and CYP2E1 preparations. Trabectedin 0-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 12235454-1 2002 OBJECTIVES: Our objective was to investigate the effects of genetic polymorphisms of cytochrome P450 (CYP) 2C9 on the pharmacokinetics of glyburide (INN, glibenclamide) and glimepiride, two widely used sulfonylurea antidiabetic drugs. Glyburide 154-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-110 12235454-5 2002 However, in individuals heterozygous for the CYP2C9*3 allele, the median total area under the plasma concentration-time curve of glyburide (n = 2) was 280% (P < or = .05) and that of glimepiride (n = 3) was 267% (P < or = .01) of the respective values in subjects with the CYP2C9*1/*1 genotype (n = 5 and n = 12, respectively). Glyburide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 12235454-5 2002 However, in individuals heterozygous for the CYP2C9*3 allele, the median total area under the plasma concentration-time curve of glyburide (n = 2) was 280% (P < or = .05) and that of glimepiride (n = 3) was 267% (P < or = .01) of the respective values in subjects with the CYP2C9*1/*1 genotype (n = 5 and n = 12, respectively). Glyburide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 279-285 12235454-5 2002 However, in individuals heterozygous for the CYP2C9*3 allele, the median total area under the plasma concentration-time curve of glyburide (n = 2) was 280% (P < or = .05) and that of glimepiride (n = 3) was 267% (P < or = .01) of the respective values in subjects with the CYP2C9*1/*1 genotype (n = 5 and n = 12, respectively). glimepiride 186-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 12698985-3 2002 MATERIALS AND METHODS: We chose 3 model drugs that are metabolized by distinct cytochrome P450 (CYP) isoforms (theophylline, phenytoin and cyclosporine for CYPIA2, CYP2C9/2C19 and CYP3A4, respectively). Cyclosporine 139-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 12360109-0 2002 Contributions of CYP2D6, CYP2C9 and CYP2C19 to the biotransformation of E- and Z-doxepin in healthy volunteers. e- and z-doxepin 72-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 12360109-2 2002 We studied the effects of genetic polymorphisms in CYP2D6, CYP2C9 and CYP2C19 on E- and Z-doxepin pharmacokinetics in humans. e- and z-doxepin 81-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 12360109-8 2002 E-doxepin oral clearance was also significantly lower in carriers of CYP2C9*3/*3 (238 l h(-1) ). Doxepin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 12235454-7 2002 CONCLUSIONS: Genetic polymorphisms of CYP2C9 markedly affect the pharmacokinetics of both glyburide and glimepiride. Glyburide 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 12235454-7 2002 CONCLUSIONS: Genetic polymorphisms of CYP2C9 markedly affect the pharmacokinetics of both glyburide and glimepiride. glimepiride 104-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 12235454-8 2002 The influence of the CYP2C9*3 variant allele on glyburide and glimepiride pharmacokinetics may be clinically significant. Glyburide 48-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 12359989-1 2002 BACKGROUND: The cytochrome P450 CYP2C9 enzyme (CYP2C9) metabolizes many clinically important drugs, for example, phenytoin, warfarin and the angiotensin II type 1 (AT(1)) receptor antagonists, losartan and irbesartan. Irbesartan 206-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 12359989-3 2002 The CYP2C9 genotype is known to determine sensitivity to and dose requirements for both warfarin and phenytoin, and also the rate of metabolism of losartan. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12359989-3 2002 The CYP2C9 genotype is known to determine sensitivity to and dose requirements for both warfarin and phenytoin, and also the rate of metabolism of losartan. Phenytoin 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12359989-3 2002 The CYP2C9 genotype is known to determine sensitivity to and dose requirements for both warfarin and phenytoin, and also the rate of metabolism of losartan. Losartan 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 12359989-5 2002 OBJECTIVE: To determine whether the CYP2C9genotype influences the blood pressure-decreasing response to antihypertensive treatment with irbesartan. Irbesartan 136-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12359989-9 2002 RESULTS: The diastolic blood pressure (DBP) response differed in relation to the CYP2C9 genotype in patients given irbesartan: the reduction in patients with genotype CYP2C9*1/CYP2C9*1 (n = 33) was 7.5% and that with CYP2C9*1/CYP2C9*2 (n = 12) was 14.4% ( P= 0.036). Irbesartan 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 12359989-9 2002 RESULTS: The diastolic blood pressure (DBP) response differed in relation to the CYP2C9 genotype in patients given irbesartan: the reduction in patients with genotype CYP2C9*1/CYP2C9*1 (n = 33) was 7.5% and that with CYP2C9*1/CYP2C9*2 (n = 12) was 14.4% ( P= 0.036). Irbesartan 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 12359989-9 2002 RESULTS: The diastolic blood pressure (DBP) response differed in relation to the CYP2C9 genotype in patients given irbesartan: the reduction in patients with genotype CYP2C9*1/CYP2C9*1 (n = 33) was 7.5% and that with CYP2C9*1/CYP2C9*2 (n = 12) was 14.4% ( P= 0.036). Irbesartan 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 12359989-9 2002 RESULTS: The diastolic blood pressure (DBP) response differed in relation to the CYP2C9 genotype in patients given irbesartan: the reduction in patients with genotype CYP2C9*1/CYP2C9*1 (n = 33) was 7.5% and that with CYP2C9*1/CYP2C9*2 (n = 12) was 14.4% ( P= 0.036). Irbesartan 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 12359989-9 2002 RESULTS: The diastolic blood pressure (DBP) response differed in relation to the CYP2C9 genotype in patients given irbesartan: the reduction in patients with genotype CYP2C9*1/CYP2C9*1 (n = 33) was 7.5% and that with CYP2C9*1/CYP2C9*2 (n = 12) was 14.4% ( P= 0.036). Irbesartan 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 12359989-12 2002 CONCLUSIONS: The CYP2C9 genotype seems to predict the DBP response to irbesartan, but not to atenolol, in patients with essential hypertension. Irbesartan 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 12167559-9 2002 LY333531 and N-desmethyl LY333531 were also examined for their ability to inhibit metabolism mediated by CYP2D6, CYP2C9, CYP3A, and CYP1A2. ruboxistaurin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12167559-9 2002 LY333531 and N-desmethyl LY333531 were also examined for their ability to inhibit metabolism mediated by CYP2D6, CYP2C9, CYP3A, and CYP1A2. 18-(methylaminomethyl)-17-oxa-4,14,21-triazahexacyclo[19.6.1.17,14.02,6.08,13.022,27]nonacosa-1(28),2(6),7(29),8,10,12,22,24,26-nonaene-3,5-dione 13-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12167570-11 2002 Thus, a phenolic group seems essential for efficient ortho-hydroxylation, forming catechol-M and tris-OH-M. Inhibition studies with HLM and P450s indicate that CYP2C9 and likely 2C19 are catalysts of methoxychlor-mono-demethylation. catechol 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 12167570-11 2002 Thus, a phenolic group seems essential for efficient ortho-hydroxylation, forming catechol-M and tris-OH-M. Inhibition studies with HLM and P450s indicate that CYP2C9 and likely 2C19 are catalysts of methoxychlor-mono-demethylation. tris-oh 97-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 12181452-3 2002 This report examines the regulation of CYP2C9 with respect to two specific receptors thought to be involved in phenobarbital (PB) induction, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR). Phenobarbital 111-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 12235454-8 2002 The influence of the CYP2C9*3 variant allele on glyburide and glimepiride pharmacokinetics may be clinically significant. glimepiride 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 12235444-1 2002 BACKGROUND: Phenytoin, a cytochrome P450 (CYP) 2C9 substrate, has a narrow therapeutic index and nonlinear pharmacokinetics. Phenytoin 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-50 12235444-2 2002 Therefore there is the potential for significant concentration-related adverse effects when phenytoin is coadministered with other CYP2C9 substrates. Phenytoin 92-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 12235444-3 2002 Losartan, an antihypertensive agent, is also a substrate for CYP2C9. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12235444-10 2002 In the 14 CYP2C9*1/*1 subjects, the mean AUC(0-24) of losartan was increased by 29% (284 +/- 84 ng x h/mL versus 402 +/- 128 ng x h/mL; P =.008). Losartan 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 12235444-12 2002 CONCLUSIONS: Losartan, a CYP2C9 substrate, had no effect on the pharmacokinetics of phenytoin. Losartan 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 12235444-13 2002 However, phenytoin inhibited the CYP2C9-mediated conversion of losartan to E3174. Phenytoin 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12235444-13 2002 However, phenytoin inhibited the CYP2C9-mediated conversion of losartan to E3174. Losartan 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12235444-13 2002 However, phenytoin inhibited the CYP2C9-mediated conversion of losartan to E3174. losartan carboxylic acid 75-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 12235454-1 2002 OBJECTIVES: Our objective was to investigate the effects of genetic polymorphisms of cytochrome P450 (CYP) 2C9 on the pharmacokinetics of glyburide (INN, glibenclamide) and glimepiride, two widely used sulfonylurea antidiabetic drugs. Glyburide 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-110 12181452-3 2002 This report examines the regulation of CYP2C9 with respect to two specific receptors thought to be involved in phenobarbital (PB) induction, the constitutive androstane receptor (CAR) and pregnane X receptor (PXR). Phenobarbital 126-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 12181452-11 2002 In summary, a new CAR/PXR binding site was identified in the CYP2C9 promoter, and this site seems to constitutively regulate transcription via a CAR-dependent mechanism; however, it could not be shown to account for PB inducibility of the gene. Phenobarbital 216-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12207635-0 2002 The influence of CYP2B6, CYP2C9 and CYP2D6 genotypes on the formation of the potent antioestrogen Z-4-hydroxy-tamoxifen in human liver. z-4-hydroxy-tamoxifen 98-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 12207635-1 2002 AIMS: To investigate in a large panel of 50 human liver samples the contribution of CYP2C9, CYP2D6, and CYP3A4 to the overall formation of the potent antioestrogen Z-4-hydroxy-tamoxifen, and how various genotypes affect its formation from tamoxifen. z-4-hydroxy-tamoxifen 164-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 12207635-5 2002 RESULTS: Z-4-hydroxy-tamoxifen was formed by supersomes expressing CYP2B6, CYP2C9, CYP2C19 and CYP2D6, but not CYP3A4. z-4-hydroxy-tamoxifen 9-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 12207635-7 2002 Furthermore, Z-4-hydroxy-tamoxifen formation significantly correlated with both CYP2C9 expression (r(s)=0.256, P<0.05) and CYP2D6 expression (r(s)=0.309, P<0.05). z-4-hydroxy-tamoxifen 13-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 12207635-9 2002 CYP2D6 and CYP2C9 contribute on average 45 and 46%, respectively, to the overall formation of Z-4-hydroxy-tamoxifen. z-4-hydroxy-tamoxifen 94-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 12207635-10 2002 CONCLUSIONS: CYP2B6, CYP2C9 and CYP2D6 genotypes all affected Z-4-hydroxy-tamoxifen formation and can predict individual ability to catalyse this reaction. z-4-hydroxy-tamoxifen 62-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 12207635-1 2002 AIMS: To investigate in a large panel of 50 human liver samples the contribution of CYP2C9, CYP2D6, and CYP3A4 to the overall formation of the potent antioestrogen Z-4-hydroxy-tamoxifen, and how various genotypes affect its formation from tamoxifen. Tamoxifen 176-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 12207639-0 2002 Intra-individual variability in urinary losartan oxidation ratio, an in vivo marker of CYP2C9 activity. Losartan 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 12207635-2 2002 METHODS: The formation of Z-4-hydroxy-tamoxifen from 10 microm tamoxifen was studied in human liver microsomes (n=50), characterized for CYP2B6, CYP2C9, CYP2D6 and CYP3A4 expression, and CYP2B6, CYP2C9 and CYP2D6 genotype. z-4-hydroxy-tamoxifen 26-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 12124312-5 2002 For the 4"-hydroxylation of diclofenac, CYP2C9 had a lower K(m) and a higher V(max) than CYP2C19. Diclofenac 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 11991950-1 2002 The fully active dihydroxylated metabolite of vitamin D(3) induces the expression of CYP3A4 and, to a lesser extent, CYP2B6 and CYP2C9 genes in normal differentiated primary human hepatocytes. Vitamin D 46-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 12130704-5 2002 Parallel increases in CYP2C mRNAs, measured by Northern blotting and/or RNase protection, were found in rifampicin-treated hepatocytes, with CYP2C8, CYP2C9, and CYP2C19 transcripts exhibiting increases of 688 +/- 635, 207 +/- 49, and 230 +/- 60%, respectively, versus an 8.8-fold enhancement of CYP3A4 mRNA levels. Rifampin 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 12130704-7 2002 Phenobarbital (100 microM) was a powerful inducer of CYP2C9 (850%) and CYP2C19 (735%) mRNA content, and also increased CYP2C8 (610%) and CYP3A4 (205%) transcripts. Phenobarbital 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 12123234-0 2002 Leu208Val and Ile181Leu variants of cytochrome P450 CYP2C9 are not related to the acenocoumarol dose requirement in a Spanish population. ile181leu 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 12152005-0 2002 Enantiospecific effects of cytochrome P450 2C9 amino acid variants on ibuprofen pharmacokinetics and on the inhibition of cyclooxygenases 1 and 2. Ibuprofen 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-46 12065445-11 2002 The present study demonstrates that CYP3A4 is the principal enzyme responsible for ketamine N-demethylation in human liver microsomes and that CYP2B6 and CYP2C9 have a minor contribution to ketamine N-demethylation at therapeutic concentrations of the drug. ketamine n 190-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 12136253-11 2002 The application of the relative substrate-activity factor (RSF)-approach and the calculation of the contribution of various isoforms in the ifosfamide 4-hydroxylation yielded the following results: CYP 3A4: 58+/-31%, CYP 2A6: 25+/-15%, and CYP 2C9: 5+/-2% of the total measured 4-hydroxylation. Ifosfamide 140-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-247 12136253-12 2002 A correlation between 4-hydroxylation and the N-dechloroethylation rates of ifosfamide and the activities of isoenzymes indicates the involvement of both CYP 3A4 ( P=0.026) and CYP 2C9 ( P=0.012) in the 4-hydroxylation reaction and of CYP 3A4 ( P<0.01) in the N-dechloroethylation reaction. Ifosfamide 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-184 12065442-4 2002 The reactions examined were CYP2C9-catalyzed diclofenac 4"-hydroxylation, CYP2D6-catalyzed dextromethorphan O-demethylation and thioridazine S-oxidation, CYP2C19-catalyzed imipramine N-demethylation, CYP3A4-catalyzed midazolam 1"-hydroxylation, and CYP1A2-catalyzed tacrine 1-hydroxylation. Diclofenac 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 12065445-0 2002 Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Nitrogen 55-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12065445-0 2002 Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Ketamine 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12065445-9 2002 In human lymphoblast-expressed P450, the activities of CYP2B6 were higher than those of CYP3A4 and CYP2C9 at three concentrations of ketamine, 0.005, 0.05, and 0.5 mM. Ketamine 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12065698-0 2002 Effect of albumin and cytosol on enzyme kinetics of tolbutamide hydroxylation and on inhibition of CYP2C9 by gemfibrozil in human liver microsomes. Gemfibrozil 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12152005-9 2002 Ex vivo formation of thromboxane B(2), reflecting cyclooxygenase type 1 inhibition, depended significantly on the CYP2C9 polymorphism. thromboxane b 21-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 12065698-1 2002 The effect of human serum albumin (Hsa) and human liver cytosol (Hlc) on the in vitro enzyme kinetics of the formation of hydroxytolbutamide (CYP2C9 marker reaction) and the inhibitory effect of gemfibrozil on tolbutamide hydroxylation were examined using human liver microsomes. hydroxymethyltolbutamide 122-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 12065698-1 2002 The effect of human serum albumin (Hsa) and human liver cytosol (Hlc) on the in vitro enzyme kinetics of the formation of hydroxytolbutamide (CYP2C9 marker reaction) and the inhibitory effect of gemfibrozil on tolbutamide hydroxylation were examined using human liver microsomes. Tolbutamide 129-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 12065698-6 2002 The K(i) (6 microM) of gemfibrozil for CYP2C9, calculated using total drug concentrations, was increased by Hlc (8 microM), Hsa (40 microM), or both (72 microM). Gemfibrozil 23-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 12152005-10 2002 The maximal inhibition of thromboxane B(2) formation and the area under the effect-time curve were larger in carriers of the slow CYP2C9 genotypes *1/*3, *2/*3, and *3/*3 than in *1/*1 carriers; the same trend was observed for prostaglandin E(2), reflecting cyclooxygenase type 2 inhibition. thromboxane b 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 12152005-10 2002 The maximal inhibition of thromboxane B(2) formation and the area under the effect-time curve were larger in carriers of the slow CYP2C9 genotypes *1/*3, *2/*3, and *3/*3 than in *1/*1 carriers; the same trend was observed for prostaglandin E(2), reflecting cyclooxygenase type 2 inhibition. Prostaglandins E 227-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 12180536-5 2002 Rh-CYP2C19, -CYP1A2, and -CYP2C9 were able to N-demethylate cis- and trans-doxepin. n-demethylate 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 12180536-4 2002 Furafylline (CYP1A2) and sulfaphenazole (CYP2C9) inhibited the N-demethylation to a lesser extent while quinidine (CYP2D6) or troleandomycine (CYP3A4) had no effect. Sulfaphenazole 25-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 12180536-5 2002 Rh-CYP2C19, -CYP1A2, and -CYP2C9 were able to N-demethylate cis- and trans-doxepin. cis- and trans-doxepin 60-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 12180536-4 2002 Furafylline (CYP1A2) and sulfaphenazole (CYP2C9) inhibited the N-demethylation to a lesser extent while quinidine (CYP2D6) or troleandomycine (CYP3A4) had no effect. Nitrogen 63-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 12019187-4 2002 With concentrations ranging from 50 to 500 microM, sulfamethoxazole was a selective inhibitor of CYP2C9-mediated tolbutamide hydroxylation in human liver microsomes and recombinant CYP2C9, with apparent IC(50) (K(i)) values of 544 microM (271 microM) and 456 microM, respectively. Sulfamethoxazole 51-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 12084621-3 2002 Isozymes selective for imidazolidine oxidation in order of decreasing overall activity are CYP3A4>CYP2C19 or CYP2A6>CYP2C9, while those selective for nitroimine reduction are CYP1A2, CYP2B6, CYP2D6 and CYP2E1. Imidazolidines 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 12084621-3 2002 Isozymes selective for imidazolidine oxidation in order of decreasing overall activity are CYP3A4>CYP2C19 or CYP2A6>CYP2C9, while those selective for nitroimine reduction are CYP1A2, CYP2B6, CYP2D6 and CYP2E1. nitroimine 156-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 12110367-7 2002 In reconstituted systems containing CYP2C9, CYP2C19, and CYP3A4, the formation of 3",4"-diHPPH was also enhanced by catalase to different extents. 3",4"-dihpph 82-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 12019187-0 2002 Trimethoprim and sulfamethoxazole are selective inhibitors of CYP2C8 and CYP2C9, respectively. Trimethoprim, Sulfamethoxazole Drug Combination 0-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 12019187-4 2002 With concentrations ranging from 50 to 500 microM, sulfamethoxazole was a selective inhibitor of CYP2C9-mediated tolbutamide hydroxylation in human liver microsomes and recombinant CYP2C9, with apparent IC(50) (K(i)) values of 544 microM (271 microM) and 456 microM, respectively. Sulfamethoxazole 51-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 12019187-4 2002 With concentrations ranging from 50 to 500 microM, sulfamethoxazole was a selective inhibitor of CYP2C9-mediated tolbutamide hydroxylation in human liver microsomes and recombinant CYP2C9, with apparent IC(50) (K(i)) values of 544 microM (271 microM) and 456 microM, respectively. Tolbutamide 113-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 12019187-6 2002 Based on estimated total hepatic concentrations (or free plasma concentrations) of the drugs and the scaling model, one would expect in vivo in humans 80% (26%) and 13% (24%) inhibition of the metabolic clearance of CYP2C8 and CYP2C9 substrates by trimethoprim and sulfamethoxazole, respectively. Trimethoprim 248-260 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 227-233 12019187-6 2002 Based on estimated total hepatic concentrations (or free plasma concentrations) of the drugs and the scaling model, one would expect in vivo in humans 80% (26%) and 13% (24%) inhibition of the metabolic clearance of CYP2C8 and CYP2C9 substrates by trimethoprim and sulfamethoxazole, respectively. Sulfamethoxazole 265-281 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 227-233 12019187-7 2002 In conclusion, trimethoprim and sulfamethoxazole can be used as selective inhibitors of CYP2C8 and CYP2C9 in in vitro studies. Trimethoprim, Sulfamethoxazole Drug Combination 15-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12019187-8 2002 In humans, trimethoprim and sulfamethoxazole may inhibit the activities of CYP2C8 and CYP2C9, respectively. Trimethoprim, Sulfamethoxazole Drug Combination 11-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 12160484-14 2002 Lower rates of V11294 metabolism to some V11294 metabolites were also observed with cDNA-expressed CYP2C9, CYP2C19 and CYP2D6, whereas only very low or undetectable rates of V11294 metabolism were observed with cDNA-expressed CYP1A2, CYP2A6, CYP2B6, CYP2C8 and CYP2E1. 3-(3-cyclopentyloxy-4-methoxybenzyl)-6-ethylamino-8-isopropyl-3H-purine 15-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12081638-3 2002 The aim of this pilot study was to clarify if PR has an effect on losartan oxidation used as a measure of CYP2C9 activity. Losartan 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 12160484-14 2002 Lower rates of V11294 metabolism to some V11294 metabolites were also observed with cDNA-expressed CYP2C9, CYP2C19 and CYP2D6, whereas only very low or undetectable rates of V11294 metabolism were observed with cDNA-expressed CYP1A2, CYP2A6, CYP2B6, CYP2C8 and CYP2E1. 3-(3-cyclopentyloxy-4-methoxybenzyl)-6-ethylamino-8-isopropyl-3H-purine 41-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 12044584-4 2002 In contrast, fluvastatin (mainly metabolized by CYP 2C9) and pravastatin (eliminated by other metabolic routes) are less subject to this interaction. Fluvastatin 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-55 11867622-5 2002 The specific CYP 2C9 inhibitor, sulfaphenazole, prevented both the enhanced cell proliferation and up-regulation of cyclin D1. Sulfaphenazole 32-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-20 11867622-10 2002 Overexpression of CYP 2C9 significantly increased the expression of MKP-1, as did incubation with 11,12-EET. 11,12-epoxy-5,8,14-eicosatrienoic acid 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-25 11955666-6 2002 CYP2C9 catalyzed the 4"-demethylation of tamarixetin, whereas CYP2D6 did not seem to play any role in the metabolism of the selected flavonoids. tamarixetin 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11950794-0 2002 Metabolism of (+)- and (-)-limonenes to respective carveols and perillyl alcohols by CYP2C9 and CYP2C19 in human liver microsomes. (+)- and (-)-limonenes 14-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 11950794-0 2002 Metabolism of (+)- and (-)-limonenes to respective carveols and perillyl alcohols by CYP2C9 and CYP2C19 in human liver microsomes. carveol 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 11950794-0 2002 Metabolism of (+)- and (-)-limonenes to respective carveols and perillyl alcohols by CYP2C9 and CYP2C19 in human liver microsomes. perillyl alcohol 64-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 11950794-4 2002 Sulfaphenazole, flavoxamine, and antibodies raised against purified liver cytochrome P450 (P450) 2C9 that inhibit both CYP2C9- and 2C19-dependent activities, significantly inhibited microsomal oxidations of (+)- and (-)-limonene enantiomers. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11950794-4 2002 Sulfaphenazole, flavoxamine, and antibodies raised against purified liver cytochrome P450 (P450) 2C9 that inhibit both CYP2C9- and 2C19-dependent activities, significantly inhibited microsomal oxidations of (+)- and (-)-limonene enantiomers. flavoxamine 16-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11950794-4 2002 Sulfaphenazole, flavoxamine, and antibodies raised against purified liver cytochrome P450 (P450) 2C9 that inhibit both CYP2C9- and 2C19-dependent activities, significantly inhibited microsomal oxidations of (+)- and (-)-limonene enantiomers. (+)- and (-)-limonene 207-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11950794-5 2002 The limonene oxidation activities correlated well with contents of CYP2C9 and activities of tolbutamide methyl hydroxylation in liver microsomes of 62 human samples, whereas these activities did not correlate with contents of CYP2C19 and activities of S-mephenytoin 4-hydroxylation. Limonene 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 11950794-8 2002 These results suggest that both (+)- and (-)-limonene enantiomers are oxidized at 6- and 7-positions by CYP2C9 and CYP2C19 in human liver microsomes. (+)- and (-)-limonene 32-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11950794-9 2002 CYP2C9 may be more important than CYP2C19 in catalyzing limonene oxidations in human liver microsomes, since levels of the former protein are more abundant than CYP2C19 in these human samples. Limonene 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11964373-2 2002 Here, we assessed the mechanisms by which the epoxyeicosatrienoic acids (EETs) generated by a CYP 2C enzyme control interendothelial gap junctional communication. epoxyeicosatrienoic acids 46-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11964373-4 2002 The initial phase was sensitive to the CYP 2C9 inhibitor sulfaphenazole and the protein kinase A (PKA) inhibitors Rp-cAMPS and KT5720 and could be mimicked by forskolin and caged cAMP as well as by the PKA activators 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole 3",5"-cyclic monophosphorothioate sodium salt and Sp-cAMPS. Sulfaphenazole 57-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-46 11964373-2 2002 Here, we assessed the mechanisms by which the epoxyeicosatrienoic acids (EETs) generated by a CYP 2C enzyme control interendothelial gap junctional communication. eets 73-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11956508-9 2002 Results of in vitro experiments showed that glyburide is metabolized by cytochrome P450 (CYP) 2C9, 2C19, and 3A4. Glyburide 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-97 11964373-4 2002 The initial phase was sensitive to the CYP 2C9 inhibitor sulfaphenazole and the protein kinase A (PKA) inhibitors Rp-cAMPS and KT5720 and could be mimicked by forskolin and caged cAMP as well as by the PKA activators 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole 3",5"-cyclic monophosphorothioate sodium salt and Sp-cAMPS. 5,6-dichloro-1-beta-d-ribofuranosylbenzimidazole 3",5"-cyclic monophosphorothioate sodium salt 217-311 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-46 11964373-4 2002 The initial phase was sensitive to the CYP 2C9 inhibitor sulfaphenazole and the protein kinase A (PKA) inhibitors Rp-cAMPS and KT5720 and could be mimicked by forskolin and caged cAMP as well as by the PKA activators 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole 3",5"-cyclic monophosphorothioate sodium salt and Sp-cAMPS. adenosine-3',5'-cyclic phosphorothioate 316-324 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-46 11956512-0 2002 Impact of CYP2C9 amino acid polymorphisms on glyburide kinetics and on the insulin and glucose response in healthy volunteers. Glyburide 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). Glyburide 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). Glyburide 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). Glyburide 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). Glyburide 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). ile359leu 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). ile359leu 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). ile359leu 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 11956512-2 2002 We hypothesized that glyburide may be a substrate of cytochrome P450 2C9 (CYP2C9), an enzyme that has two low-activity amino acid variants-Arg144Cys (CYP2C9*2) and Ile359Leu (CYP2C9*3). ile359leu 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 11956512-8 2002 RESULTS: Pharmacokinetics of glyburide depended significantly on CYP2C9 genotypes. Glyburide 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 11956512-11 2002 CONCLUSIONS: Carriers of the CYP2C9 variant *3 had decreased oral clearances of glyburide. Glyburide 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 11956512-12 2002 This confirms that glyburide is metabolized by CYP2C9. Glyburide 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 11926893-0 2002 Association between CYP2C9 genetic variants and anticoagulation-related outcomes during warfarin therapy. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 11926893-2 2002 The principal enzyme involved in warfarin metabolism is CYP2C9, and 2 relatively common variant forms with reduced activity have been identified, CYP2C9*2 and CYP2C9*3. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 11926893-2 2002 The principal enzyme involved in warfarin metabolism is CYP2C9, and 2 relatively common variant forms with reduced activity have been identified, CYP2C9*2 and CYP2C9*3. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 11926893-2 2002 The principal enzyme involved in warfarin metabolism is CYP2C9, and 2 relatively common variant forms with reduced activity have been identified, CYP2C9*2 and CYP2C9*3. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 11926893-4 2002 OBJECTIVE: To determine if CYP2C9*2 and CYP2C9*3 variants are associated with overanticoagulation and bleeding events during warfarin therapy. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11926893-4 2002 OBJECTIVE: To determine if CYP2C9*2 and CYP2C9*3 variants are associated with overanticoagulation and bleeding events during warfarin therapy. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 11926893-14 2002 CONCLUSIONS: The results of our study suggest that the CYP2C9*2 and CYP2C9*3 polymorphisms are associated with an increased risk of overanticoagulation and of bleeding events among patients in a warfarin anticoagulation clinic setting, although small numbers in some cases would suggest the need for caution in interpretation. Warfarin 195-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 11926893-14 2002 CONCLUSIONS: The results of our study suggest that the CYP2C9*2 and CYP2C9*3 polymorphisms are associated with an increased risk of overanticoagulation and of bleeding events among patients in a warfarin anticoagulation clinic setting, although small numbers in some cases would suggest the need for caution in interpretation. Warfarin 195-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11926893-15 2002 Screening for CYP2C9 variants may allow clinicians to develop dosing protocols and surveillance techniques to reduce the risk of adverse drug reactions in patients receiving warfarin. Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 11918509-5 2002 DISCUSSION: RTV is an inducer of the hepatic isoenzymes CYP1A2, CYP1A4, and CYP2C9/19 and leads to extensive metabolism of acenocoumarol that cannot be balanced by dose increases. Ritonavir 12-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 11918509-5 2002 DISCUSSION: RTV is an inducer of the hepatic isoenzymes CYP1A2, CYP1A4, and CYP2C9/19 and leads to extensive metabolism of acenocoumarol that cannot be balanced by dose increases. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 11901091-3 2002 To this end, the kinetic profiles of three model CYP2C9 substrates (flurbiprofen, naproxen, and piroxicam) were studied using purified CYP2C9*1 (wild-type) and variants involving active site amino acid changes, including the naturally occurring variants CYP2C9*3 (Leu359) and CYP2C9*5 (Glu360) and the man-made mutant CYP2C9 F114L. Flurbiprofen 68-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11901091-3 2002 To this end, the kinetic profiles of three model CYP2C9 substrates (flurbiprofen, naproxen, and piroxicam) were studied using purified CYP2C9*1 (wild-type) and variants involving active site amino acid changes, including the naturally occurring variants CYP2C9*3 (Leu359) and CYP2C9*5 (Glu360) and the man-made mutant CYP2C9 F114L. Naproxen 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11901091-3 2002 To this end, the kinetic profiles of three model CYP2C9 substrates (flurbiprofen, naproxen, and piroxicam) were studied using purified CYP2C9*1 (wild-type) and variants involving active site amino acid changes, including the naturally occurring variants CYP2C9*3 (Leu359) and CYP2C9*5 (Glu360) and the man-made mutant CYP2C9 F114L. Piroxicam 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11901091-4 2002 CYP2C9*1 (wild-type) metabolized each of the three compounds with a distinctive profile reflective of typical hyperbolic (flurbiprofen), biphasic (naproxen), and substrate inhibition (piroxicam) kinetics. Flurbiprofen 122-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-4 2002 CYP2C9*1 (wild-type) metabolized each of the three compounds with a distinctive profile reflective of typical hyperbolic (flurbiprofen), biphasic (naproxen), and substrate inhibition (piroxicam) kinetics. Naproxen 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-4 2002 CYP2C9*1 (wild-type) metabolized each of the three compounds with a distinctive profile reflective of typical hyperbolic (flurbiprofen), biphasic (naproxen), and substrate inhibition (piroxicam) kinetics. Piroxicam 184-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-5 2002 CYP2C9*3 metabolism was again hyperbolic for flurbiprofen, of a linear form for naproxen (no saturation noted), and exhibited substrate inhibition with piroxicam. Flurbiprofen 45-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-5 2002 CYP2C9*3 metabolism was again hyperbolic for flurbiprofen, of a linear form for naproxen (no saturation noted), and exhibited substrate inhibition with piroxicam. Naproxen 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-5 2002 CYP2C9*3 metabolism was again hyperbolic for flurbiprofen, of a linear form for naproxen (no saturation noted), and exhibited substrate inhibition with piroxicam. Piroxicam 152-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-6 2002 CYP2C9*5-mediated metabolism was hyperbolic for flurbiprofen and piroxicam but linear with respect to naproxen turnover. Flurbiprofen 48-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-6 2002 CYP2C9*5-mediated metabolism was hyperbolic for flurbiprofen and piroxicam but linear with respect to naproxen turnover. Piroxicam 65-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901091-6 2002 CYP2C9*5-mediated metabolism was hyperbolic for flurbiprofen and piroxicam but linear with respect to naproxen turnover. Naproxen 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11901092-4 2002 CYP2A6 (coumarin 7-hydroxylation) and CYP2C9 (diclofenac 4"-hydroxylation) activities exhibited a mixed type of inhibition comprising competitive and noncompetitive components in response to SN-38, the K(i) values being 181 and 156 microM, respectively. Diclofenac 46-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 11901092-4 2002 CYP2A6 (coumarin 7-hydroxylation) and CYP2C9 (diclofenac 4"-hydroxylation) activities exhibited a mixed type of inhibition comprising competitive and noncompetitive components in response to SN-38, the K(i) values being 181 and 156 microM, respectively. Irinotecan 191-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 24-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 35-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11901092-9 2002 These results mean that CPT-11 and SN-38 interact with human P450 isoforms, such as CYP2A6, CYP2C9, and CYP3A4, in vitro and imply that the significant drug interactions involving CPT-11 may be caused by a mechanism-based inactivation of CYP3A4 by SN-38 as an active metabolite of CPT-11 rather than competitive inhibition. Irinotecan 180-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 11910269-6 2002 Because paroxetine is a mild inhibitor of CYP1A2, CYP2C9, CYP2C19, and CYP3A4, increase of (S)-methadone concentrations in both EMs and PMs could be mediated by inhibition of any of these isozymes. Paroxetine 8-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 11854151-6 2002 Ketoconazole seemed to be the most selective for CYP3A4, although it also inhibited CYP2C9. Ketoconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 11956677-0 2002 Inhibition by atovaquone of CYP2C9-mediated sulphamethoxazole hydroxylamine formation. Atovaquone 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11956677-0 2002 Inhibition by atovaquone of CYP2C9-mediated sulphamethoxazole hydroxylamine formation. sulfamethoxazole hydroxylamine 44-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11956677-2 2002 METHODS: Generation of SMX-HA from SMX was measured directly using high-performance liquid chromatography in human liver microsomes or expressed CYP2C9*1, with or without preincubation with reduced nicotinamide adenine dinucleotide phosphate, and the inhibition constant (K(i)) for atovaquone was determined. sulfamethoxazole hydroxylamine 23-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 11956677-2 2002 METHODS: Generation of SMX-HA from SMX was measured directly using high-performance liquid chromatography in human liver microsomes or expressed CYP2C9*1, with or without preincubation with reduced nicotinamide adenine dinucleotide phosphate, and the inhibition constant (K(i)) for atovaquone was determined. Sulfamethoxazole 23-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 11956677-7 2002 Compared with human liver microsomes, CYP2C9*1 showed an eightfold greater specific activity for SMX-HA generation; as for liver microsomes, CYP2C9*1 activity was inhibited by atovaquone. Atovaquone 176-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 11956677-7 2002 Compared with human liver microsomes, CYP2C9*1 showed an eightfold greater specific activity for SMX-HA generation; as for liver microsomes, CYP2C9*1 activity was inhibited by atovaquone. Atovaquone 176-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 11956677-8 2002 CONCLUSIONS: Atovaquone is a relatively weak inhibitor of CYP2C9-mediated SMX-HA formation in vitro. Atovaquone 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11956677-8 2002 CONCLUSIONS: Atovaquone is a relatively weak inhibitor of CYP2C9-mediated SMX-HA formation in vitro. Sulfamethoxazole 74-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11925616-8 2002 The enzyme activity of CYP2C9 catalyzing oxidation of tolbutamide to hydroxy tolbutamide in S9 fraction of the cell was determined by high performance liquid chromatography(HPLC). Tolbutamide 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11925616-8 2002 The enzyme activity of CYP2C9 catalyzing oxidation of tolbutamide to hydroxy tolbutamide in S9 fraction of the cell was determined by high performance liquid chromatography(HPLC). hydroxymethyltolbutamide 69-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11875362-0 2002 CYP2C9 polymorphism: impact on tolbutamide pharmacokinetics and response. Tolbutamide 31-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11893129-0 2002 The frequency and effects of cytochrome P450 (CYP) 2C9 polymorphisms in patients receiving warfarin. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-54 11893129-3 2002 To better understand the interaction between genotype and response to warfarin therapy, we determined the frequency and functional effects of polymorphisms of the predominant cytochrome P450 subfamily responsible for warfarin metabolism (eg, CYP2C9) in an ethnically defined U.S. patient population. Warfarin 217-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 242-248 11893129-13 2002 CONCLUSIONS: CYP2C9 polymorphisms are common, associated with significant reductions in warfarin dose, and partly account for interpatient variability in warfarin sensitivity. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 11893129-13 2002 CONCLUSIONS: CYP2C9 polymorphisms are common, associated with significant reductions in warfarin dose, and partly account for interpatient variability in warfarin sensitivity. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 11907487-12 2002 Consistent with expression data, formation of verapamil metabolites catalyzed by CYP3A4 and CYP2C was shown. Verapamil 46-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-97 11936218-4 2002 Oroxylin A inhibited diclofenac 4-hydroxylation (CYP2C9) activity with a IC50 of 6.7 microM. 5,7-dihydroxy-6-methoxy-2-phenylchromen-4-one 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11936218-4 2002 Oroxylin A inhibited diclofenac 4-hydroxylation (CYP2C9) activity with a IC50 of 6.7 microM. Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11875364-9 2002 Tolbutamide was confirmed as a substrate of the genetically polymorphic enzyme CYP2C9. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 11875364-0 2002 Impact of CYP2C9 and CYP2C19 polymorphisms on tolbutamide kinetics and the insulin and glucose response in healthy volunteers. Tolbutamide 46-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 11875365-0 2002 Effects of CYP2C19 and CYP2C9 genetic polymorphisms on the disposition of and blood glucose lowering response to tolbutamide in humans. Glucose 84-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11875365-0 2002 Effects of CYP2C19 and CYP2C9 genetic polymorphisms on the disposition of and blood glucose lowering response to tolbutamide in humans. Tolbutamide 113-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11875365-1 2002 Several recent in-vitro data have revealed that CYP2C19, in addition to CYP2C9, is also involved in the 4-methylhydroxylation of tolbutamide. Tolbutamide 129-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 11875365-7 2002 In subjects heterozygous for the CYP2C9*3 allele, C(max) and AUC of tolbutamide were significantly greater and the plasma half-life significantly longer than those in homozygous CYP2C9*1 subjects. Tolbutamide 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 11875365-7 2002 In subjects heterozygous for the CYP2C9*3 allele, C(max) and AUC of tolbutamide were significantly greater and the plasma half-life significantly longer than those in homozygous CYP2C9*1 subjects. Tolbutamide 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Glucose 43-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Glucose 43-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Glucose 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Glucose 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Glucose 243-250 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11875365-9 2002 The estimated AUC of the increase in serum glucose after oral intake of 100 g dextrose was 2.7-fold higher in subjects with the wild-type CYP2C9 genotype than in those with CYP2C9*1/*3, but CYP2C19 genetic polymorphism did not alter the blood glucose lowering effect of tolbutamide. Tolbutamide 270-281 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11875364-1 2002 Tolbutamide is known to be metabolized by cytochrome P450 2C9 (CYP2C9), and the effects of the CYP2C9 amino acid polymorphisms *2 (Arg144Cys) and *3 (Ile359Leu) could be important for drug treatment with tolbutamide and for use of tolbutamide as a CYP2C9 test drug. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-61 11875365-11 2002 Our results strongly suggest that the disposition and hypoglycemic effect of tolbutamide are affected mainly by CYP2C9 genetic polymorphism, but not by CYP2C19 polymorphism. Tolbutamide 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 11875365-13 2002 This suggests that, at least in vivo, tolbutamide remains a selective probe for measuring CYP2C9 activity in humans. Tolbutamide 38-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 11875364-1 2002 Tolbutamide is known to be metabolized by cytochrome P450 2C9 (CYP2C9), and the effects of the CYP2C9 amino acid polymorphisms *2 (Arg144Cys) and *3 (Ile359Leu) could be important for drug treatment with tolbutamide and for use of tolbutamide as a CYP2C9 test drug. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 11853173-3 2002 Additionally, nonylphenol competitively inhibited diclofenac 4"-hydroxylation by CYP2C9 and S-mephenytoin 4"-hydroxylation by CYP2C19 with Ki values of 5.3 and 37 microM, respectively. nonylphenol 14-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11853173-3 2002 Additionally, nonylphenol competitively inhibited diclofenac 4"-hydroxylation by CYP2C9 and S-mephenytoin 4"-hydroxylation by CYP2C19 with Ki values of 5.3 and 37 microM, respectively. diclofenac 4" 50-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11853173-2 2002 When determined at 2 mM substrate concentration, nonylphenol (1 mM) most efficiently inhibited aminopyrine N-demethylation by CYP2C9 and CYP2C19, by 61% and 59%, respectively, followed by CYP2D6, CYP1A2, CYP2C18 and CYP2C8 (46-51%), whereas inhibition of the activities by other CYPs was less than 27%. nonylphenol 49-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 11853173-5 2002 These results suggest that nonylphenol inhibits human hepatic CYPs, especially CYP2C9 and CYP2C19, and steroidogenic CYP17 activities. nonylphenol 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 11936702-2 2002 METHODS: Bufuralol 1"-hydroxylation and tolbutamide 4-methylhydroxylation were used as index reactions for CYP2D6 and CYP2C9, respectively. bufuralol 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 11853173-2 2002 When determined at 2 mM substrate concentration, nonylphenol (1 mM) most efficiently inhibited aminopyrine N-demethylation by CYP2C9 and CYP2C19, by 61% and 59%, respectively, followed by CYP2D6, CYP1A2, CYP2C18 and CYP2C8 (46-51%), whereas inhibition of the activities by other CYPs was less than 27%. aminopyrine n 95-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 11816012-7 2002 Since celecoxib should be metabolized primarily by cytochrome 2C9 (CYP2C9), a poor metabolizer (PM) for this cytochrome P450 enzyme was included in the study. Celecoxib 6-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 11792677-9 2002 Using human microsomes, tibolone, 3alpha-hydroxy tibolone, 3beta-hydroxy tibolone, and Delta(4)-tibolone appeared to be at least 50-fold less potent inhibitors of CYP1A2, CYP2C9, CYP2E1, and CYP3A4 compared with enzyme-selective inhibitors. tibolone 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 11792677-9 2002 Using human microsomes, tibolone, 3alpha-hydroxy tibolone, 3beta-hydroxy tibolone, and Delta(4)-tibolone appeared to be at least 50-fold less potent inhibitors of CYP1A2, CYP2C9, CYP2E1, and CYP3A4 compared with enzyme-selective inhibitors. 3-hydroxytibolone 34-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 11792677-9 2002 Using human microsomes, tibolone, 3alpha-hydroxy tibolone, 3beta-hydroxy tibolone, and Delta(4)-tibolone appeared to be at least 50-fold less potent inhibitors of CYP1A2, CYP2C9, CYP2E1, and CYP3A4 compared with enzyme-selective inhibitors. 3-hydroxytibolone 59-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 11792677-9 2002 Using human microsomes, tibolone, 3alpha-hydroxy tibolone, 3beta-hydroxy tibolone, and Delta(4)-tibolone appeared to be at least 50-fold less potent inhibitors of CYP1A2, CYP2C9, CYP2E1, and CYP3A4 compared with enzyme-selective inhibitors. delta(4)-tibolone 87-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 11936702-7 2002 CONCLUSION: All five H1-antihistamines studied inhibited CYP2D6 markedly, but only cyclizine and promethazine inhibited CYP2C9 at concentrations above that usually seen in plasma. Cyclizine 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 11936702-7 2002 CONCLUSION: All five H1-antihistamines studied inhibited CYP2D6 markedly, but only cyclizine and promethazine inhibited CYP2C9 at concentrations above that usually seen in plasma. Promethazine 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 11875991-0 2002 Warfarin therapy in a patient homozygous for the CYP2C9 3 allele. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11876575-6 2002 Fluvoxamine is a potent CYP1A2 and CYP2C19 inhibitor, and a moderate CYP2C9, CYP2D6, and CYP3A4 inhibitor. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 11925850-0 2002 [Relationship of polymorphism in CYP2C9 to genetic susceptibility to diclofenac-induced influenza-virus-associated encephalopathy]. Diclofenac 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 11831544-1 2002 Modafinil has been reported to produce a concentration-related suppression of CYP2C9 activity in vitro in primary cultures of human hepatocytes. Modafinil 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 11831544-5 2002 Since (S)-warfarin is predominantly metabolized via CYP2C9, the results indicate that the marked suppression of CYP2C9 activity in vitro does not translate into a similar effect clinically. Warfarin 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11831544-5 2002 Since (S)-warfarin is predominantly metabolized via CYP2C9, the results indicate that the marked suppression of CYP2C9 activity in vitro does not translate into a similar effect clinically. Warfarin 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 11925850-2 2002 CYP2C9 is known as the major cytochrome P450 gene product that catalyzes diclofenac in human liver. Diclofenac 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11925850-6 2002 We suggest that this mutation in the CYP2C9 gene may be related to diclofenac-induced influenza-virus-associated encephalopathy. Diclofenac 67-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11823761-0 2002 Pharmacokinetics of losartan and its metabolite E-3174 in relation to the CYP2C9 genotype. Losartan 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 11679585-3 2002 In previous work, we have shown that CYP2C9 is inducible in primary human hepatocytes by xenobiotics including dexamethasone, rifampicin, and phenobarbital. Dexamethasone 111-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11679585-3 2002 In previous work, we have shown that CYP2C9 is inducible in primary human hepatocytes by xenobiotics including dexamethasone, rifampicin, and phenobarbital. Rifampin 126-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11679585-3 2002 In previous work, we have shown that CYP2C9 is inducible in primary human hepatocytes by xenobiotics including dexamethasone, rifampicin, and phenobarbital. Phenobarbital 142-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11679585-8 2002 Identification of these functional elements provides rational mechanistic basis for CYP2C9 induction by dexamethasone (submicromolar concentrations), and by phenobarbital and rifampicin, respectively. Dexamethasone 104-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 11679585-8 2002 Identification of these functional elements provides rational mechanistic basis for CYP2C9 induction by dexamethasone (submicromolar concentrations), and by phenobarbital and rifampicin, respectively. Phenobarbital 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 11679585-8 2002 Identification of these functional elements provides rational mechanistic basis for CYP2C9 induction by dexamethasone (submicromolar concentrations), and by phenobarbital and rifampicin, respectively. Rifampin 175-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 11753007-2 2002 Both parecoxib and valdecoxib inhibit human cytochrome P450 2C9 (CYP2C9) activity in vitro. parecoxib 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-63 11753007-2 2002 Both parecoxib and valdecoxib inhibit human cytochrome P450 2C9 (CYP2C9) activity in vitro. parecoxib 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 11753007-2 2002 Both parecoxib and valdecoxib inhibit human cytochrome P450 2C9 (CYP2C9) activity in vitro. valdecoxib 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-63 11753007-2 2002 Both parecoxib and valdecoxib inhibit human cytochrome P450 2C9 (CYP2C9) activity in vitro. valdecoxib 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 11753007-3 2002 Thus, a potential exists for in vivo interactions with other CYP2C9 substrates, including propofol. Propofol 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 12405866-17 2002 Sulfonylurea drugs utilise mainly CYP2C9 for metabolism, and this isoenzyme may be induced by ritonavir and nelfinavir with a resulting decrease in efficacy of the sulfonylurea. Sulfonylurea Compounds 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 12405866-17 2002 Sulfonylurea drugs utilise mainly CYP2C9 for metabolism, and this isoenzyme may be induced by ritonavir and nelfinavir with a resulting decrease in efficacy of the sulfonylurea. Sulfonylurea Compounds 164-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 12405866-18 2002 Losartan may have increased effect when coadministered with ritonavir and nelfinavir because of the induction of CYP2C9 and the expected increase in formation of the active metabolite, E-3174. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12405866-18 2002 Losartan may have increased effect when coadministered with ritonavir and nelfinavir because of the induction of CYP2C9 and the expected increase in formation of the active metabolite, E-3174. Ritonavir 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 12405866-18 2002 Losartan may have increased effect when coadministered with ritonavir and nelfinavir because of the induction of CYP2C9 and the expected increase in formation of the active metabolite, E-3174. Nelfinavir 74-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 11823761-0 2002 Pharmacokinetics of losartan and its metabolite E-3174 in relation to the CYP2C9 genotype. losartan carboxylic acid 48-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 11823761-1 2002 BACKGROUND AND AIM: Losartan is metabolized by polymorphic CYP2C9 to E-3174. Losartan 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 11823761-2 2002 Our aim was to evaluate the pharmacokinetics of losartan and E-3174 in relation to the CYP2C9 genotype. Losartan 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 11823761-2 2002 Our aim was to evaluate the pharmacokinetics of losartan and E-3174 in relation to the CYP2C9 genotype. losartan carboxylic acid 61-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 11823761-3 2002 METHODS: A 50-mg oral dose of losartan was given to 22 Swedish volunteers with different CYP2C9 genotypes. Losartan 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 11888331-15 2002 Inhibition of the CYP2C9 and CYP3A isoenzymes by zafirlukast has been reported in vitro. zafirlukast 49-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 11823761-6 2002 RESULTS: The maximum plasma concentration of E-3174 was significantly (P <.05) lower in the CYP2C9*1/*3 (n = 5) and CYP2C9*2/*3 (n = 4) groups compared with the CYP2C9*1/*1 (n = 6) and CYP2C9*1/*2 (n = 3) groups and extremely low in 1 subject with the CYP2C9*3/*3 genotype. losartan carboxylic acid 45-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 11823761-6 2002 RESULTS: The maximum plasma concentration of E-3174 was significantly (P <.05) lower in the CYP2C9*1/*3 (n = 5) and CYP2C9*2/*3 (n = 4) groups compared with the CYP2C9*1/*1 (n = 6) and CYP2C9*1/*2 (n = 3) groups and extremely low in 1 subject with the CYP2C9*3/*3 genotype. losartan carboxylic acid 45-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11823761-6 2002 RESULTS: The maximum plasma concentration of E-3174 was significantly (P <.05) lower in the CYP2C9*1/*3 (n = 5) and CYP2C9*2/*3 (n = 4) groups compared with the CYP2C9*1/*1 (n = 6) and CYP2C9*1/*2 (n = 3) groups and extremely low in 1 subject with the CYP2C9*3/*3 genotype. losartan carboxylic acid 45-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11823761-6 2002 RESULTS: The maximum plasma concentration of E-3174 was significantly (P <.05) lower in the CYP2C9*1/*3 (n = 5) and CYP2C9*2/*3 (n = 4) groups compared with the CYP2C9*1/*1 (n = 6) and CYP2C9*1/*2 (n = 3) groups and extremely low in 1 subject with the CYP2C9*3/*3 genotype. losartan carboxylic acid 45-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11823761-6 2002 RESULTS: The maximum plasma concentration of E-3174 was significantly (P <.05) lower in the CYP2C9*1/*3 (n = 5) and CYP2C9*2/*3 (n = 4) groups compared with the CYP2C9*1/*1 (n = 6) and CYP2C9*1/*2 (n = 3) groups and extremely low in 1 subject with the CYP2C9*3/*3 genotype. losartan carboxylic acid 45-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 11823761-7 2002 The ratio of the total losartan area under the plasma concentration-time curve (AUC) to the total E-3174 AUC (AUC(losartan)/AUC(E-3174)) was higher in the subject with the CYP2C9*3/*3 genotype (30-fold) and also in the CYP2C9*1/*3 and *2/*3 groups (approximately 2- and 3-fold, respectively) compared with the CYP2C9*1/*1 group. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 11823761-7 2002 The ratio of the total losartan area under the plasma concentration-time curve (AUC) to the total E-3174 AUC (AUC(losartan)/AUC(E-3174)) was higher in the subject with the CYP2C9*3/*3 genotype (30-fold) and also in the CYP2C9*1/*3 and *2/*3 groups (approximately 2- and 3-fold, respectively) compared with the CYP2C9*1/*1 group. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 11823761-7 2002 The ratio of the total losartan area under the plasma concentration-time curve (AUC) to the total E-3174 AUC (AUC(losartan)/AUC(E-3174)) was higher in the subject with the CYP2C9*3/*3 genotype (30-fold) and also in the CYP2C9*1/*3 and *2/*3 groups (approximately 2- and 3-fold, respectively) compared with the CYP2C9*1/*1 group. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 11823761-7 2002 The ratio of the total losartan area under the plasma concentration-time curve (AUC) to the total E-3174 AUC (AUC(losartan)/AUC(E-3174)) was higher in the subject with the CYP2C9*3/*3 genotype (30-fold) and also in the CYP2C9*1/*3 and *2/*3 groups (approximately 2- and 3-fold, respectively) compared with the CYP2C9*1/*1 group. Losartan 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 11823761-10 2002 CONCLUSION: The CYP2C9*3 allele was shown to be associated with decreased formation of E-3174 from losartan. losartan carboxylic acid 87-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 11823761-10 2002 CONCLUSION: The CYP2C9*3 allele was shown to be associated with decreased formation of E-3174 from losartan. Losartan 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 11823761-11 2002 The significant differences between genotypes in plasma and urine losartan/E-3174 ratios and the good correlation between the plasma and urine ratios suggest that the losartan/E-3174 ratio in 0- to 8-hour urine specimens may serve as a phenotyping assay for CYP2C9 activity. Losartan 167-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 258-264 15618707-5 2002 Addition of HSA resulted in smaller Km,u values for diclofenac and terfenadine metabolism by CYP2C9 and CYP3A4, respectively, and the Ki,u value for ketoconazole inhibition of CYP3A4 was also reduced. Diclofenac 52-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15618695-7 2002 In contrast, the inhibitory effects of phenacetin, diclofenac, S-mephenytoin, dextromethorphan, bufuralol and terfenadine, typical substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, respectively, on each recombinant CYP activity decreased after preincubation. Phenacetin 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 12036392-9 2002 The CYP3A family metabolises lovastatin, simvastatin, atorvastatin and cerivastatin, whereas CYP2C9 metabolises fluvastatin. Fluvastatin 112-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 11794436-6 2002 Genetic analysis of the cytochrome P450 (CYP) isoenzyme 2C9, which mediates the major deactivating pathway of S-warfarin, revealed that the patient was a compound heterozygote carrying two variant alleles: CYP2C9*2 and CYP2C9*3. Warfarin 110-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 15618705-11 2002 CYP2C enzymes were suggested to play major roles in metabolizing (+)-carveol to (+)-carvone and (+)-carvone to (+)-carveol by liver microsomes, since the activities were inhibited significantly by anti-human CYP2C9 antibodies in these animal species. carveol 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 15618705-11 2002 CYP2C enzymes were suggested to play major roles in metabolizing (+)-carveol to (+)-carvone and (+)-carvone to (+)-carveol by liver microsomes, since the activities were inhibited significantly by anti-human CYP2C9 antibodies in these animal species. carvone 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 15618705-11 2002 CYP2C enzymes were suggested to play major roles in metabolizing (+)-carveol to (+)-carvone and (+)-carvone to (+)-carveol by liver microsomes, since the activities were inhibited significantly by anti-human CYP2C9 antibodies in these animal species. carvone 96-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 15618705-11 2002 CYP2C enzymes were suggested to play major roles in metabolizing (+)-carveol to (+)-carvone and (+)-carvone to (+)-carveol by liver microsomes, since the activities were inhibited significantly by anti-human CYP2C9 antibodies in these animal species. carveol 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 15618705-12 2002 Studies with recombinant P450 enzymes suggested that CYP2C9 and 2C19 in humans and CYP2C11 in untreated male rats were the major enzymes in metabolizing (+)-carvone. carvone 153-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 15618706-5 2002 CYP2C9 expressed in human B-lymphoblastoid cells catalyzed efficiently the MALDO activity for 11-oxo-Delta(8)-tetrahydrocannabinol (10.1 nmol/min/nmol P450), while the catalytic activities of other human CYPs expressed in the cells were lesser extents. 11-oxo-delta(8)-tetrahydrocannabinol 94-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 15618706-7 2002 These results indicate that CYP2C9 and CYP3A4 are major enzymes responsible for the MALDO activity in human liver for 11-oxo-Delta(8)-tetrahydrocannabinol and 9-anthraldehyde, respectively. 11-oxo-delta(8)-tetrahydrocannabinol 118-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 15618706-7 2002 These results indicate that CYP2C9 and CYP3A4 are major enzymes responsible for the MALDO activity in human liver for 11-oxo-Delta(8)-tetrahydrocannabinol and 9-anthraldehyde, respectively. 9-anthraldehyde 159-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 15618707-2 2002 The effect of addition of rat liver cytosol to an in vitro system using human liver microsomes was examined by measuring the catalytic activities of CYP2C9 (tolbutamide and diclofenac) and CYP3A4 (terfenadine). Tolbutamide 157-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 15618707-4 2002 After addition of rat liver cytosol, the unbound Km value (Km,u) for terfenadine metabolism by CYP3A4, and the unbound Ki value of miconazole (Ki,u) for CYP2C9 were smaller than for the controls. Miconazole 131-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 15618707-5 2002 Addition of HSA resulted in smaller Km,u values for diclofenac and terfenadine metabolism by CYP2C9 and CYP3A4, respectively, and the Ki,u value for ketoconazole inhibition of CYP3A4 was also reduced. Terfenadine 67-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 15618695-7 2002 In contrast, the inhibitory effects of phenacetin, diclofenac, S-mephenytoin, dextromethorphan, bufuralol and terfenadine, typical substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, respectively, on each recombinant CYP activity decreased after preincubation. bufuralol 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 15618695-7 2002 In contrast, the inhibitory effects of phenacetin, diclofenac, S-mephenytoin, dextromethorphan, bufuralol and terfenadine, typical substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, respectively, on each recombinant CYP activity decreased after preincubation. Terfenadine 110-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 11794436-6 2002 Genetic analysis of the cytochrome P450 (CYP) isoenzyme 2C9, which mediates the major deactivating pathway of S-warfarin, revealed that the patient was a compound heterozygote carrying two variant alleles: CYP2C9*2 and CYP2C9*3. Warfarin 110-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 11794436-7 2002 The patient"s enhanced sensitivity to warfarin 5 mg/day can be ascribed to decreased clearance of S-warfarin secondary to genetic alteration of the gene encoding CYP2C9, resulting in a life-threatening complication. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11794436-7 2002 The patient"s enhanced sensitivity to warfarin 5 mg/day can be ascribed to decreased clearance of S-warfarin secondary to genetic alteration of the gene encoding CYP2C9, resulting in a life-threatening complication. Sulfur 98-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11794436-7 2002 The patient"s enhanced sensitivity to warfarin 5 mg/day can be ascribed to decreased clearance of S-warfarin secondary to genetic alteration of the gene encoding CYP2C9, resulting in a life-threatening complication. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11717174-3 2001 At higher concentrations, DDB marginally inhibited caffeine N(3)-demethylation (CYP1A2), diclofenac 4"-hydroxylation (CYP2C9), and dextromethorphan O-demethylation (CYP2D6) activities, but this compound had no effect on CYP2A6-, CYP2C19-, and CYP2E1-mediated reactions. ddb 26-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 11724084-9 2001 DISCUSSION: Phenytoin is principally metabolized by CYP2C9. Phenytoin 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11708911-6 2001 The selectivity analysis singled out CYP2C8 as the most different of the four CYP2C enzymes with amino acids with distinct properties in positions 114, 205, and 476 (Ser, Phe, and Ile, respectively) compared to the other enzymes. Serine 166-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-42 11708911-6 2001 The selectivity analysis singled out CYP2C8 as the most different of the four CYP2C enzymes with amino acids with distinct properties in positions 114, 205, and 476 (Ser, Phe, and Ile, respectively) compared to the other enzymes. Phenylalanine 171-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-42 11708911-6 2001 The selectivity analysis singled out CYP2C8 as the most different of the four CYP2C enzymes with amino acids with distinct properties in positions 114, 205, and 476 (Ser, Phe, and Ile, respectively) compared to the other enzymes. Isoleucine 180-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-42 11708911-7 2001 An inverse pharmacophore model for CYP2C9 was constructed from the selective regions, and the model agreed with the docking of diclofenac where the properties of the ligand overlapped with the pharmacophoric points in the model. Diclofenac 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 11829203-0 2001 The role of CYP2C9 genotype in the metabolism of diclofenac in vivo and in vitro. Diclofenac 49-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Warfarin 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-56 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Warfarin 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Acenocoumarol 127-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-56 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Acenocoumarol 127-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Phenytoin 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-56 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Phenytoin 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Tolbutamide 153-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-56 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Tolbutamide 153-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Losartan 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-56 11829203-1 2001 INTRODUCTION: The polymorphic cytochrome P450 enzyme 2C9 (CYP2C9) catalyses the metabolism of many drugs including S-warfarin, acenocoumarol, phenytoin, tolbutamide, losartan and most of the non-steroidal anti-inflammatory drugs. Losartan 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11829203-2 2001 Diclofenac is metabolised to 4"-hydroxy (OH), the major diclofenac metabolite, 3"-OH and 3"-OH-4"-methoxy metabolites by CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 11829203-2 2001 Diclofenac is metabolised to 4"-hydroxy (OH), the major diclofenac metabolite, 3"-OH and 3"-OH-4"-methoxy metabolites by CYP2C9. 4-hydroxyphenylacetic acid 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 11829203-2 2001 Diclofenac is metabolised to 4"-hydroxy (OH), the major diclofenac metabolite, 3"-OH and 3"-OH-4"-methoxy metabolites by CYP2C9. 3"-oh 79-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 11829203-2 2001 Diclofenac is metabolised to 4"-hydroxy (OH), the major diclofenac metabolite, 3"-OH and 3"-OH-4"-methoxy metabolites by CYP2C9. 3"-oh-4" 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 11829203-3 2001 The aim of the present study was to clarify the impact of the CYP2C9 polymorphism on the metabolism of diclofenac both in vivo and in vitro. Diclofenac 103-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 11714868-5 2001 The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Oligonucleotides 4-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 11714868-5 2001 The oligonucleotide-based array made it possible to detect different levels of induction within the CYP2C family, with rifampin causing a 6.5-fold increase in expression of CYP2C8 and a 3.7-fold increase in CYP2C9 while having no effect on the level of CYP2C18 mRNA. Rifampin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 11740344-0 2001 Identification of a null allele of CYP2C9 in an African-American exhibiting toxicity to phenytoin. Phenytoin 88-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Phenytoin 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Phenytoin 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Glipizide 155-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Glipizide 155-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Tolbutamide 170-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 11740344-2 2001 Two polymorphic alleles CYP2C9*2 and CYP2C9*3 have been documented which affect the metabolism and clinical toxicity of drugs such as phenytoin, warfarin, glipizide, and tolbutamide. Tolbutamide 170-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11740344-9 2001 Sequencing studies revealed that the individual was homozygous for a new CYP2C9 allele (CYP2C9*6) with the deletion of an adenine at base pair 818 of the cDNA. Adenine 122-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 11740344-9 2001 Sequencing studies revealed that the individual was homozygous for a new CYP2C9 allele (CYP2C9*6) with the deletion of an adenine at base pair 818 of the cDNA. Adenine 122-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 11740344-12 2001 The study also demonstrates the severe clinical consequences to patients with a null mutation in CYP2C9 after treatment with normal doses of phenytoin. Phenytoin 141-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 11736863-0 2001 In vitro evaluation of valproic acid as an inhibitor of human cytochrome P450 isoforms: preferential inhibition of cytochrome P450 2C9 (CYP2C9). Valproic Acid 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-134 11736863-0 2001 In vitro evaluation of valproic acid as an inhibitor of human cytochrome P450 isoforms: preferential inhibition of cytochrome P450 2C9 (CYP2C9). Valproic Acid 23-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 11736863-3 2001 RESULTS: Valproic acid competitively inhibited CYP2C9 activity with a Ki value of 600 microM. Valproic Acid 9-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 11736863-7 2001 CONCLUSIONS: Valproic acid inhibits the activity of CYP2C9 at clinically relevant concentrations in human liver microsomes. Valproic Acid 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11736863-8 2001 Inhibition of CYP2C9 can explain some of the effects of valproic acid on the pharmacokinetics of other drugs, such as phenytoin. Valproic Acid 56-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 11736863-8 2001 Inhibition of CYP2C9 can explain some of the effects of valproic acid on the pharmacokinetics of other drugs, such as phenytoin. Phenytoin 118-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 11736863-9 2001 Co-administration of high doses of valproic acid with drugs that are primarily metabolized by CYP2C9 may result in significant drug interactions. Valproic Acid 35-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11594733-0 2001 Interaction of new sulfaphenazole derivatives with human liver cytochrome p450 2Cs: structural determinants required for selective recognition by CYP 2C9 and for inhibition of human CYP 2Cs. Sulfaphenazole 19-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-153 11719730-1 2001 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug and a substrate of cytochrome P4502C9 (CYP2C9). Gemfibrozil 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-200 11719730-1 2001 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug and a substrate of cytochrome P4502C9 (CYP2C9). glimepiride 113-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-200 11719730-1 2001 OBJECTIVE: Our objective was to study the effects of gemfibrozil on the pharmacokinetics and pharmacodynamics of glimepiride, a new sulfonylurea antidiabetic drug and a substrate of cytochrome P4502C9 (CYP2C9). glimepiride 113-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 202-208 11692083-0 2001 Early acenocoumarol overanticoagulation among cytochrome P450 2C9 poor metabolizers. Acenocoumarol 6-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 11692083-1 2001 Cytochrome P450 2C9 (CYP2C9) is the enzyme that terminates the anticoagulant effect of warfarin. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 11692083-1 2001 Cytochrome P450 2C9 (CYP2C9) is the enzyme that terminates the anticoagulant effect of warfarin. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 11692083-2 2001 The heterozygous carriers of the two allelic variants CYP2C9*2 and CYP2C9*3 have been associated with impaired warfarin metabolism and a higher risk of haemorrhage. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 11692083-2 2001 The heterozygous carriers of the two allelic variants CYP2C9*2 and CYP2C9*3 have been associated with impaired warfarin metabolism and a higher risk of haemorrhage. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 11692083-3 2001 Only three CYP2C9 poor metabolizers (CYP2C9*3/CYP2C9*3) initiating warfarin treatment have so far been identified, all of them with a dramatic overdose occurring a few days after treatment initiation. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 11588061-0 2001 Genetic polymorphism in exon 4 of cytochrome P450 CYP2C9 may be associated with warfarin sensitivity in Chinese patients. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 11588061-1 2001 CYP2C9 polymorphisms reported in Caucasians (Arg144Cys in exon 3 and Ile359Leu in exon 7) are extremely uncommon in Chinese persons. ile359leu 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11588061-2 2001 The genotype of CYP2C9 in this population was characterized to investigate its relation with the interindividual variation in warfarin dosages. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 11602509-0 2001 Gemfibrozil is a potent inhibitor of human cytochrome P450 2C9. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 11602509-3 2001 Gemfibrozil strongly and competitively inhibited CYP2C9 activity, with a K(i) (IC(50)) value of 5.8 (9.6) microM. Gemfibrozil 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Gemfibrozil 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Glyburide 212-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 11602509-7 2001 In conclusion, gemfibrozil inhibits the activity of CYP2C9 at clinically relevant concentrations, and this is the likely mechanism by which gemfibrozil interacts with CYP2C9 substrate drugs, such as warfarin and glyburide. Glyburide 212-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 11692083-3 2001 Only three CYP2C9 poor metabolizers (CYP2C9*3/CYP2C9*3) initiating warfarin treatment have so far been identified, all of them with a dramatic overdose occurring a few days after treatment initiation. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11692083-3 2001 Only three CYP2C9 poor metabolizers (CYP2C9*3/CYP2C9*3) initiating warfarin treatment have so far been identified, all of them with a dramatic overdose occurring a few days after treatment initiation. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 11692083-4 2001 Acenocoumarol, another coumarinic anticoagulant, has recently been shown to be metabolized by CYP2C9. Acenocoumarol 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. ferric sulfate 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-31 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. ferric sulfate 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. ferric sulfate 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. galactose-1-phosphate 45-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-31 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. galactose-1-phosphate 45-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 11594733-3 2001 This indicates that the CYP 2C9-iron(III)-NH(2)R bond previously described to exist in the CYP 2C9-SPA complex does not play a key role in the high affinity of SPA for CYP 2C9. galactose-1-phosphate 45-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-98 11580286-10 2001 The effects of ticlopidine on CYP 2C19 are very analogous with those previously described for the inactivation of CYP 2C9 by tienilic acid. Ticlopidine 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-121 11580286-10 2001 The effects of ticlopidine on CYP 2C19 are very analogous with those previously described for the inactivation of CYP 2C9 by tienilic acid. Ticrynafen 125-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-121 11580286-11 2001 This suggests that a similar electrophilic intermediate, possibly a thiophene S-oxide, is involved in the inactivation of CYP 2C19 and CYP 2C9 by ticlopidine and tienilic acid, respectively. thiophene s-oxide 68-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-142 11580286-11 2001 This suggests that a similar electrophilic intermediate, possibly a thiophene S-oxide, is involved in the inactivation of CYP 2C19 and CYP 2C9 by ticlopidine and tienilic acid, respectively. Ticlopidine 146-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-142 11580286-11 2001 This suggests that a similar electrophilic intermediate, possibly a thiophene S-oxide, is involved in the inactivation of CYP 2C19 and CYP 2C9 by ticlopidine and tienilic acid, respectively. Ticrynafen 162-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-142 11560876-8 2001 Finally, 10 microM CCM induced both CYP2C9 and CYP2B6, strengthening the evidence that hPXR is involved in the regulation of these genes. Fruitcal 19-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 11673755-0 2001 Severe phenytoin intoxication in a subject homozygous for CYP2C9*3. Phenytoin 7-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11673755-5 2001 In the absence of any acquired predisposing factor for phenytoin toxicity, genetic mutations in the cytochrome P450 (CYP) enzymes responsible for phenytoin metabolism (CYP2C9 and CYP2C19) were suspected. Phenytoin 146-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 11673755-7 2001 In view of the markedly reduced metabolic activity of CYP2C*3 in comparison with the wild-type enzyme (about one fifth) and of the minor role of CYP2C19 in phenytoin metabolism, it is likely that CYP2C9*3 mutation was largely responsible for drug overdose. Phenytoin 156-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 11560876-0 2001 Calcium channel modulators of the dihydropyridine family are human pregnane X receptor activators and inducers of CYP3A, CYP2B, and CYP2C in human hepatocytes. 1,4-dihydropyridine 34-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-137 11907638-1 2001 BACKGROUND: The concurrent use of amiodarone and warfarin inhibits metabolism of S-warfarinby cytochrome P450 (CYP) 2C9, thereby increasing the anticoagulant effect of warfarin. Amiodarone 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-119 11678778-11 2001 CYP2C9 metabolizes a wide variety of drugs including the anticoagulant warfarin, antidiabetic agents such as tolbutamide, anticonvulsants such as phenytoin, and nonsteroidal anti-inflammatory drugs. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11678778-11 2001 CYP2C9 metabolizes a wide variety of drugs including the anticoagulant warfarin, antidiabetic agents such as tolbutamide, anticonvulsants such as phenytoin, and nonsteroidal anti-inflammatory drugs. Tolbutamide 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11678778-14 2001 Severe and life-threatening bleeding episodes have been reported in CYP2C9 PMs exposed to warfarin. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11907638-6 2001 CONCLUSIONS: It was suggested that inhibition of CYP2C9 by desethylamiodarone, the active metabolite of amiodarone, plays an important role in the interaction of warfarin and amiodarone. Amiodarone 104-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11907638-1 2001 BACKGROUND: The concurrent use of amiodarone and warfarin inhibits metabolism of S-warfarinby cytochrome P450 (CYP) 2C9, thereby increasing the anticoagulant effect of warfarin. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-119 11907638-1 2001 BACKGROUND: The concurrent use of amiodarone and warfarin inhibits metabolism of S-warfarinby cytochrome P450 (CYP) 2C9, thereby increasing the anticoagulant effect of warfarin. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-119 11907638-2 2001 Amiodarone primarily inhibits CYP1A2 and CYP3A4, and desethylamiodarone primarily inhibits CYP2C9. desethylamiodarone 53-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 11907638-6 2001 CONCLUSIONS: It was suggested that inhibition of CYP2C9 by desethylamiodarone, the active metabolite of amiodarone, plays an important role in the interaction of warfarin and amiodarone. desethylamiodarone 59-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11907638-6 2001 CONCLUSIONS: It was suggested that inhibition of CYP2C9 by desethylamiodarone, the active metabolite of amiodarone, plays an important role in the interaction of warfarin and amiodarone. Amiodarone 67-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11907638-6 2001 CONCLUSIONS: It was suggested that inhibition of CYP2C9 by desethylamiodarone, the active metabolite of amiodarone, plays an important role in the interaction of warfarin and amiodarone. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11668218-0 2001 Phenytoin metabolic ratio: a putative marker of CYP2C9 activity in vivo. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 11601668-6 2001 Celecoxib is metabolized by CYP2C9 and may be increased or decreased by CYP2C9 modifiers. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11601668-6 2001 Celecoxib is metabolized by CYP2C9 and may be increased or decreased by CYP2C9 modifiers. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 11668218-8 2001 The CYP2C9-mediated production of (S)-p-HPPH represented the major metabolic pathway of phenytoin biotransformation as its excretion accounted for 95.6 + 0.9% of "total" p-HPPH excretion over the 96 h collection interval. Phosphorus 34-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 11668218-8 2001 The CYP2C9-mediated production of (S)-p-HPPH represented the major metabolic pathway of phenytoin biotransformation as its excretion accounted for 95.6 + 0.9% of "total" p-HPPH excretion over the 96 h collection interval. Phenytoin 88-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 11668218-11 2001 CYP2C9 genotyping and several phenytoin metabolic indices are correlated with CYP2C9 activity in vivo. Phenytoin 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 11668218-12 2001 The utility of phenytoin to predict the metabolism of other CYP2C9 substrates justifies further evaluation. Phenytoin 15-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 11557918-9 2001 CONCLUSION: The presence of even one copy of CYP2C9*3 reduces profoundly the metabolic clearance of S -acenocoumarol. Acenocoumarol 100-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 11558570-3 2001 CYP2C18 catalyzed BPA metabolism most efficiently, followed by CYP2C19 and CYP2C9. bisphenol A 18-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 11558570-4 2001 CYP2C9 and CYP2C18 exhibited the highest affinity (Km=3.9 microM) for BPA metabolism. bisphenol A 70-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11557918-0 2001 Altered pharmacokinetics of R- and S-acenocoumarol in a subject heterozygous for CYP2C9*3. r- 28-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11557918-0 2001 Altered pharmacokinetics of R- and S-acenocoumarol in a subject heterozygous for CYP2C9*3. Acenocoumarol 35-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11551516-6 2001 Importantly, we show elevated levels of CYP2C9 protein to be associated with cadmium accumulation in liver. Cadmium 77-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 11551516-8 2001 One is that variation in CYP2C9 protein levels may be, in part, attributed to an individual"s non-workplace exposure to cadmium, or an individual"s CYP2C9 genotype may be a risk factor for cadmium accumulation. Cadmium 120-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11551516-8 2001 One is that variation in CYP2C9 protein levels may be, in part, attributed to an individual"s non-workplace exposure to cadmium, or an individual"s CYP2C9 genotype may be a risk factor for cadmium accumulation. Cadmium 189-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11551516-8 2001 One is that variation in CYP2C9 protein levels may be, in part, attributed to an individual"s non-workplace exposure to cadmium, or an individual"s CYP2C9 genotype may be a risk factor for cadmium accumulation. Cadmium 189-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 11470765-9 2001 The association of the CYP2C9 polymorphism to colorectal cancer risk could be related to CYP2C9-mediated metabolic activation of polycyclic aromatic hydrocarbons and heterocyclic aromatic amines. Polycyclic Aromatic Hydrocarbons 129-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11583336-0 2001 Reply to rebuttal: Gene variants of the cytochrome P450 CYP2C9 affect oral anticoagulation with warfarin. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 11470765-9 2001 The association of the CYP2C9 polymorphism to colorectal cancer risk could be related to CYP2C9-mediated metabolic activation of polycyclic aromatic hydrocarbons and heterocyclic aromatic amines. Polycyclic Aromatic Hydrocarbons 129-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 11470765-9 2001 The association of the CYP2C9 polymorphism to colorectal cancer risk could be related to CYP2C9-mediated metabolic activation of polycyclic aromatic hydrocarbons and heterocyclic aromatic amines. heterocyclic aromatic amines 166-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11470765-9 2001 The association of the CYP2C9 polymorphism to colorectal cancer risk could be related to CYP2C9-mediated metabolic activation of polycyclic aromatic hydrocarbons and heterocyclic aromatic amines. heterocyclic aromatic amines 166-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 11503010-1 2001 BACKGROUND: Interindividual variability in responses to warfarin is attributed to dietary vitamin K, drug interactions, age, or genetic polymorphism in the cytochrome P4502C9 enzyme (CYP2C9) (allelic variants 2C9*2 and 2C9*3 ) linked with impaired metabolism of the potent enantiomere S-warfarin. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-181 11503010-1 2001 BACKGROUND: Interindividual variability in responses to warfarin is attributed to dietary vitamin K, drug interactions, age, or genetic polymorphism in the cytochrome P4502C9 enzyme (CYP2C9) (allelic variants 2C9*2 and 2C9*3 ) linked with impaired metabolism of the potent enantiomere S-warfarin. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 11503010-1 2001 BACKGROUND: Interindividual variability in responses to warfarin is attributed to dietary vitamin K, drug interactions, age, or genetic polymorphism in the cytochrome P4502C9 enzyme (CYP2C9) (allelic variants 2C9*2 and 2C9*3 ) linked with impaired metabolism of the potent enantiomere S-warfarin. Warfarin 287-295 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-181 11503010-1 2001 BACKGROUND: Interindividual variability in responses to warfarin is attributed to dietary vitamin K, drug interactions, age, or genetic polymorphism in the cytochrome P4502C9 enzyme (CYP2C9) (allelic variants 2C9*2 and 2C9*3 ) linked with impaired metabolism of the potent enantiomere S-warfarin. Warfarin 287-295 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 11457649-0 2001 Minimal in vivo activation of CYP2C9-mediated flurbiprofen metabolism by dapsone. Flurbiprofen 46-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 11454729-5 2001 PEITC noncompetitively inhibited S-warfarin 7-hydroxylase activity catalyzed by CYP2C9 (K(i) = 6.5 +/- 0.9 microM), S-mephenytoin 4"-hydroxylase activity catalyzed by CYP2C19 (K(i) = 12.0 +/- 3.2 microM), bufuralol 1"-hydroxylase activity catalyzed by CYP2D6 (K(i) = 28.4 +/- 7.9 microM), and chlorzoxazone 6-hydroxylase activity catalyzed by CYP2E1 (K(i) = 21.5 +/- 3.4 microM). phenethyl isothiocyanate 0-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 11457649-0 2001 Minimal in vivo activation of CYP2C9-mediated flurbiprofen metabolism by dapsone. Dapsone 73-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 11457649-1 2001 Dapsone has been shown to activate flurbiprofen 4"-hydroxylation by expressed CYP2C9 enzyme and in human liver microsomes. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 11457649-1 2001 Dapsone has been shown to activate flurbiprofen 4"-hydroxylation by expressed CYP2C9 enzyme and in human liver microsomes. Flurbiprofen 35-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 11457649-9 2001 These dapsone plasma concentrations were within the range of concentrations producing activation of flurbiprofen metabolism by CYP2C9 in vitro. Dapsone 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 11457649-9 2001 These dapsone plasma concentrations were within the range of concentrations producing activation of flurbiprofen metabolism by CYP2C9 in vitro. Flurbiprofen 100-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 11452703-1 2001 Evidence for the selectivity of S-warfarin metabolism by CYP2C9 is substantial, suggesting that warfarin may be a potential CYP2C9 phenotyping probe. Warfarin 32-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 11455026-7 2001 The major effect of the Asp360Glu mutation was to increase the K(m) value relative to that of CYP2C9*1 for all three substrates: 12-fold higher for (S)-warfarin 7-hydroxylation, 5-fold higher for the 4"-hydroxylation of diclofenac, and 3-fold higher for the omega-1 hydroxylation of lauric acid. lauric acid 283-294 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11408373-6 2001 In comparison to the CYP2C9.1 variant, oxidation of losartan was significantly reduced in yeast expressing the rare CYP2C9.2 or CYP2C9.3 variants. Losartan 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 11408373-7 2001 Moreover, the rate of losartan oxidation was lower in liver microsomes from individuals hetero- or homozygous for the CYP2C9*3 allele, or homozygous for the CYP2C9*2 allele. Losartan 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 11408373-7 2001 Moreover, the rate of losartan oxidation was lower in liver microsomes from individuals hetero- or homozygous for the CYP2C9*3 allele, or homozygous for the CYP2C9*2 allele. Losartan 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 11408373-9 2001 In summary, these in vitro results indicate that CYP2C9 is the major human P450 isoenzyme responsible for losartan oxidation and that the CYP2C9 genotype contributes to interindividual differences in losartan oxidation and activation. Losartan 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11408373-9 2001 In summary, these in vitro results indicate that CYP2C9 is the major human P450 isoenzyme responsible for losartan oxidation and that the CYP2C9 genotype contributes to interindividual differences in losartan oxidation and activation. Losartan 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 11408373-9 2001 In summary, these in vitro results indicate that CYP2C9 is the major human P450 isoenzyme responsible for losartan oxidation and that the CYP2C9 genotype contributes to interindividual differences in losartan oxidation and activation. Losartan 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11455026-7 2001 The major effect of the Asp360Glu mutation was to increase the K(m) value relative to that of CYP2C9*1 for all three substrates: 12-fold higher for (S)-warfarin 7-hydroxylation, 5-fold higher for the 4"-hydroxylation of diclofenac, and 3-fold higher for the omega-1 hydroxylation of lauric acid. Warfarin 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11455026-7 2001 The major effect of the Asp360Glu mutation was to increase the K(m) value relative to that of CYP2C9*1 for all three substrates: 12-fold higher for (S)-warfarin 7-hydroxylation, 5-fold higher for the 4"-hydroxylation of diclofenac, and 3-fold higher for the omega-1 hydroxylation of lauric acid. Diclofenac 220-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 11408370-0 2001 Dapsone activation of CYP2C9-mediated metabolism: evidence for activation of multiple substrates and a two-site model. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 11408370-1 2001 Dapsone activates CYP2C9-mediated metabolism in various expression systems and is itself metabolized by CYP2C9 to its hydroxylamine metabolite. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 11408370-1 2001 Dapsone activates CYP2C9-mediated metabolism in various expression systems and is itself metabolized by CYP2C9 to its hydroxylamine metabolite. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11408370-1 2001 Dapsone activates CYP2C9-mediated metabolism in various expression systems and is itself metabolized by CYP2C9 to its hydroxylamine metabolite. Hydroxylamine 118-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11408370-4 2001 Dapsone increased the Michaelis-Menten-derived V(max) of flurbiprofen 4"-hydroxylation from 12.6 to 20.6 pmol/min/pmol P450, and lowered its K(m) from 28.9 to 10.0 microM, suggesting that dapsone activates CYP2C9-mediated flurbiprofen metabolism without displacing flurbiprofen from the active site, supporting a two-site model describing activation. Dapsone 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 11408370-4 2001 Dapsone increased the Michaelis-Menten-derived V(max) of flurbiprofen 4"-hydroxylation from 12.6 to 20.6 pmol/min/pmol P450, and lowered its K(m) from 28.9 to 10.0 microM, suggesting that dapsone activates CYP2C9-mediated flurbiprofen metabolism without displacing flurbiprofen from the active site, supporting a two-site model describing activation. Flurbiprofen 57-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 11408370-9 2001 Overall, these results suggest that dapsone activates the metabolism of multiple substrates of CYP2C9 by binding within the active site and causing positive cooperativity, thus lending further support to a two-site binding model of P450-mediated metabolism. Dapsone 36-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 11408373-0 2001 Role of CYP2C9 polymorphism in losartan oxidation. Losartan 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 11408373-2 2001 The aim of the present investigation was to study the contribution of CYP2C9 and CYP3A4 in losartan oxidation in vitro and to evaluate the role of CYP2C9 polymorphism. Losartan 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 11452703-1 2001 Evidence for the selectivity of S-warfarin metabolism by CYP2C9 is substantial, suggesting that warfarin may be a potential CYP2C9 phenotyping probe. Warfarin 32-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 11452703-1 2001 Evidence for the selectivity of S-warfarin metabolism by CYP2C9 is substantial, suggesting that warfarin may be a potential CYP2C9 phenotyping probe. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 11452703-1 2001 Evidence for the selectivity of S-warfarin metabolism by CYP2C9 is substantial, suggesting that warfarin may be a potential CYP2C9 phenotyping probe. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 11452703-4 2001 The objective of this study was to investigate the pharmacokinetics and pharmacodynamics of S- and R-warfarin in plasma following the administration of warfarin alone versus warfarin and vitamin K in CYP2C9*1 homozygotes. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 200-206 11452703-4 2001 The objective of this study was to investigate the pharmacokinetics and pharmacodynamics of S- and R-warfarin in plasma following the administration of warfarin alone versus warfarin and vitamin K in CYP2C9*1 homozygotes. Vitamin K 187-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 200-206 11452703-14 2001 Warfarin 10 mg may be safely used as a CYP2C9 probe in *1 homozygotes when given concomitantly with 10 mg of oral vitamin K. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 11353758-5 2001 Among the 12 cDNA-expressed CYP enzymes examined, CYP2B6, CYP2C9, and CYP3A4 showed high activities for the N-demethylation of both enantiomers at the substrate concentration of 1 mM. Nitrogen 15-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11408373-5 2001 Sulfaphenazole, a CYP2C9 inhibitor, blocked the formation of E-3174 at low losartan concentrations (<1 microM), whereas the inhibitory effect of triacetyloleandomycin, a CYP3A4 inhibitor, was significant only at high concentrations of losartan (>25 microM). Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 11408373-5 2001 Sulfaphenazole, a CYP2C9 inhibitor, blocked the formation of E-3174 at low losartan concentrations (<1 microM), whereas the inhibitory effect of triacetyloleandomycin, a CYP3A4 inhibitor, was significant only at high concentrations of losartan (>25 microM). losartan carboxylic acid 61-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 11408373-6 2001 In comparison to the CYP2C9.1 variant, oxidation of losartan was significantly reduced in yeast expressing the rare CYP2C9.2 or CYP2C9.3 variants. Losartan 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 11408373-6 2001 In comparison to the CYP2C9.1 variant, oxidation of losartan was significantly reduced in yeast expressing the rare CYP2C9.2 or CYP2C9.3 variants. Losartan 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 11353758-7 2001 Also, the intrinsic clearance (CL(int): V(max)/K(m)) of CYP2B6 for the N-demethylation of both enantiomers were 7 to 13 times higher than those of CYP2C9 and CYP3A4. Nitrogen 71-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 11353758-8 2001 Orphenadrine (CYP2B6 inhibitor, 500 microM) and sulfaphenazole (CYP2C9 inhibitor, 100 microM) inhibited the N-demethylase activities for both enantiomers (5 microM) in human liver microsomes by 60 to 70%, whereas cyclosporin A (CYP3A4 inhibitor, 100 microM) failed to inhibit these activities. Sulfaphenazole 48-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 11325819-0 2001 CYP2C9 and UGT1A6 genotypes modulate the protective effect of aspirin on colon adenoma risk. Aspirin 62-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11353749-10 2001 Moreover, etoricoxib (0.1-100 microM) was found to be a relatively weak inhibitor (IC(50) > 100 microM) of multiple P450s (CYP1A2, CYP2D6, CYP3A, CYP2E1, CYP2C9, and CYP2C19) in human liver microsomes. Etoricoxib 10-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 11353755-7 2001 SCH 66712 also inhibited HL microsomal CYP3A4, CYP2C9, and CYP2C19; however, the concentrations required to inhibit those isoforms were 5- to 10-fold higher than those required to inhibit CYP2D6. SCH 66712 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 11356927-3 2001 R-Norfluoxetine formation rates in a characterized microsomal bank correlated with the catalytic activities for cytochrome P450 (CYP) 2D6, CYP2C9, and CYP2C8. (R)-Norfluoxetine 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 11356927-4 2001 Expressed CYP2C9, CYP2C19, and CYP2D6 formed R-norfluoxetine following incubation with 1 microM R-fluoxetine and exhibited apparent K(m) values of 9.7, 8.5, and 1.8 microM, respectively. (R)-Norfluoxetine 45-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 11356927-4 2001 Expressed CYP2C9, CYP2C19, and CYP2D6 formed R-norfluoxetine following incubation with 1 microM R-fluoxetine and exhibited apparent K(m) values of 9.7, 8.5, and 1.8 microM, respectively. (R)-Fluoxetine 96-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 11356927-5 2001 Multivariate correlation analysis identified CYP2C9 and CYP2D6 as significant regressors with R-norfluoxetine formation. (R)-Norfluoxetine 94-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 11406737-12 2001 The mechanism underlying the interaction between rifampin and glyburide is probably induction of either CYP2C9 or P-glycoprotein or both. Rifampin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11406737-12 2001 The mechanism underlying the interaction between rifampin and glyburide is probably induction of either CYP2C9 or P-glycoprotein or both. Glyburide 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11406737-13 2001 Induction of CYP2C9 would explain the increased systemic elimination of glipizide. Glipizide 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 11356927-9 2001 Thus, it would appear that both CYP2D6 and CYP2C9 contribute to the formation of R-norfluoxetine, whereas only CYP2D6 is responsible for the conversion to S-norfluoxetine. (R)-Norfluoxetine 81-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11356927-10 2001 Since the enantiomers of fluoxetine and norfluoxetine are inhibitors of CYP2D6, upon chronic dosing, the CYP2D6-mediated metabolism of the fluoxetine enantiomers would likely be inhibited, resulting in R-norfluoxetine formation being mediated by CYP2C9 and S-norfluoxetine formation being mediated by multiple high K(m) enzymes. Fluoxetine 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 246-252 11356927-10 2001 Since the enantiomers of fluoxetine and norfluoxetine are inhibitors of CYP2D6, upon chronic dosing, the CYP2D6-mediated metabolism of the fluoxetine enantiomers would likely be inhibited, resulting in R-norfluoxetine formation being mediated by CYP2C9 and S-norfluoxetine formation being mediated by multiple high K(m) enzymes. norfluoxetine 40-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 246-252 11356927-10 2001 Since the enantiomers of fluoxetine and norfluoxetine are inhibitors of CYP2D6, upon chronic dosing, the CYP2D6-mediated metabolism of the fluoxetine enantiomers would likely be inhibited, resulting in R-norfluoxetine formation being mediated by CYP2C9 and S-norfluoxetine formation being mediated by multiple high K(m) enzymes. Fluoxetine 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 246-252 11434505-0 2001 The effect of genetic polymorphism of cytochrome P450 CYP2C9 on phenytoin dose requirement. Phenytoin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 11434505-1 2001 The cytochrome P450 enzyme CYP2C9 catalyses the metabolism of numerous therapeutic agents, including the anti-epileptic drug phenytoin. Phenytoin 125-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11434505-4 2001 Because this may be relevant in treatment with phenytoin, we studied the effect of CYP2C9 genotype on phenytoin dose requirement in a group of 60 epileptic patients on long-term phenytoin therapy. Phenytoin 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 11434505-4 2001 Because this may be relevant in treatment with phenytoin, we studied the effect of CYP2C9 genotype on phenytoin dose requirement in a group of 60 epileptic patients on long-term phenytoin therapy. Phenytoin 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 11434505-6 2001 For patients carrying at least one mutant CYP2C9 allele (n = 17), the mean phenytoin dose required to achieve a therapeutic serum concentration was about 37% lower than the mean dose required by wild-type individuals (199 mg/day versus 314 mg/day; P < 0.01). Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 11434505-8 2001 The results show that there is a strong association between CYP2C9 allelic variants and phenytoin dose requirement. Phenytoin 88-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 11325819-3 2001 NSAIDs, particularly aspirin, are glucuronidated by UGT1A6 and some classes of NSAIDs are also metabolized by cytochrome P450 (CYP) 2C9. Aspirin 21-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-135 11325819-8 2001 However, this association was absent in aspirin users who carried the CYP2C9 variant alleles (OR, 0.88; 95% CI, 0.51-1.53) or who were homozygous wild-type UGT1A6 (OR, 0.86; 95% CI, 0.50-1.50). Aspirin 40-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 11309547-11 2001 This was probably caused by inhibition of the cytochrome P-450 2C9-mediated biotransformation of glimepiride by fluconazole. glimepiride 97-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-66 11573629-7 2001 These results indicate that only CYP3A4, but not CYP2B6 or CYP2C, is involved in the metabolism of midazolam in vitro. Midazolam 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-64 11309547-11 2001 This was probably caused by inhibition of the cytochrome P-450 2C9-mediated biotransformation of glimepiride by fluconazole. Fluconazole 112-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-66 11372587-1 2001 OBJECTIVES: To determine the relative contribution of cytochromes P450 (CYP) 2C9 and 2C19 to the formation of 5-(-4-hydroxyphenyl)-5-phenylhydantion (HPPH) from phenytoin (PPH). 5-(-4-hydroxyphenyl)-5-phenylhydantion 110-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-89 11255075-2 2001 The mechanism of this drug-drug interaction has been attributed to inhibition of CYP2C9-catalyzed hydroxylation of phenytoin to its major oxidative metabolite in humans, para-hydroxyphenyl phenyl hydantoin (HPPH). Phenytoin 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11255075-2 2001 The mechanism of this drug-drug interaction has been attributed to inhibition of CYP2C9-catalyzed hydroxylation of phenytoin to its major oxidative metabolite in humans, para-hydroxyphenyl phenyl hydantoin (HPPH). para-hydroxyphenyl phenyl hydantoin 170-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11372587-1 2001 OBJECTIVES: To determine the relative contribution of cytochromes P450 (CYP) 2C9 and 2C19 to the formation of 5-(-4-hydroxyphenyl)-5-phenylhydantion (HPPH) from phenytoin (PPH). 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a 150-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-89 11255075-2 2001 The mechanism of this drug-drug interaction has been attributed to inhibition of CYP2C9-catalyzed hydroxylation of phenytoin to its major oxidative metabolite in humans, para-hydroxyphenyl phenyl hydantoin (HPPH). 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a 207-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 11372587-1 2001 OBJECTIVES: To determine the relative contribution of cytochromes P450 (CYP) 2C9 and 2C19 to the formation of 5-(-4-hydroxyphenyl)-5-phenylhydantion (HPPH) from phenytoin (PPH). Phenytoin 161-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-89 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 257-263 11372587-4 2001 Coincubation with the CYP2C9 inhibitor, sulfaphenazole (SPA), at 5 mumol/l reduced reaction velocity to less than 15% of control values. Sulfaphenazole 40-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 11372587-4 2001 Coincubation with the CYP2C9 inhibitor, sulfaphenazole (SPA), at 5 mumol/l reduced reaction velocity to less than 15% of control values. Sulfaphenazole 56-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. 4"-oh diclofenac 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. 4"-oh diclofenac 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 257-263 11465405-12 2001 While 10O microM BFBFC was metabolized to HFC by cDNA-expressed CYP3A4, little or no metabolism was observed with cDNA-expressed CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP2E1. 2,5-bis(trifluoromethyl)-7-benzyloxy-4-trifluoromethylcoumarin 17-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. Phenytoin 189-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11255075-9 2001 Diclofenac hydroxylation to 4"-OH diclofenac, a specific marker for CYP2C9 activity, was determined in HLM1-HLM6 and was highly correlated with HPPH formation in HLM1-HLM6, indicating that phenytoin hydroxylation in human liver microsomes is largely due to CYP2C9. Phenytoin 189-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 257-263 11255075-10 2001 This work presents direct evidence that the effect of fluoxetine on phenytoin blood levels may be explained by inhibition of CYP2C9-catalyzed phenytoin hydroxylation. Fluoxetine 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 11255075-10 2001 This work presents direct evidence that the effect of fluoxetine on phenytoin blood levels may be explained by inhibition of CYP2C9-catalyzed phenytoin hydroxylation. Phenytoin 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 11255075-10 2001 This work presents direct evidence that the effect of fluoxetine on phenytoin blood levels may be explained by inhibition of CYP2C9-catalyzed phenytoin hydroxylation. Phenytoin 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 11304902-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP 2C9 and 3A4. alosetron 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-97 11304903-2 2001 Alosetron is extensively metabolized by multiple cytochrome P450 (CYP) enzymes, including CYP2C9 and CYP3A4. alosetron 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 11304903-4 2001 Fluoxetine metabolism involves CYP2D6 and CYP2C9 in the formation of its major metabolite, norfluoxetine. Fluoxetine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 11304903-4 2001 Fluoxetine metabolism involves CYP2D6 and CYP2C9 in the formation of its major metabolite, norfluoxetine. norfluoxetine 91-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 11465405-19 2001 While 80 microM 7BQ was metabolized to 7HQ by cDNA-expressed CYP3A4, only low rates of metabolism were observed with cDNA-expressed CYPIA2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and CYP2E1. 7-benzyloxyquinoline 16-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 11181490-4 2001 Compounds known to activate the pregnane X receptor (PXR) such as rifampicin, or the constitutively activated receptor (CAR) such as phenobarbital, induced CYP2C8, CYP2C9, and to a lesser extent CYP2C19 mRNAs and proteins. Rifampin 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 11337938-0 2001 In-vitro metabolism of celecoxib, a cyclooxygenase-2 inhibitor, by allelic variant forms of human liver microsomal cytochrome P450 2C9: correlation with CYP2C9 genotype and in-vivo pharmacokinetics. Celecoxib 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-134 11337938-0 2001 In-vitro metabolism of celecoxib, a cyclooxygenase-2 inhibitor, by allelic variant forms of human liver microsomal cytochrome P450 2C9: correlation with CYP2C9 genotype and in-vivo pharmacokinetics. Celecoxib 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 11337938-1 2001 In-vitro studies were conducted to assess the impact of CYP2C9 genotype on the metabolism (methyl hydroxylation) and pharmacokinetics of celecoxib, a novel cyclooxygenase-2 inhibitor and CYP2C9 substrate. Celecoxib 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 11337938-2 2001 When compared to cDNA-expressed wild-type CYP2C9 (CYP2C9*1), the Vmax/Km ratio for celecoxib methyl hydroxylation was reduced by 34% and 90% in the presence of recombinant CYP2C9*2 and CYP2C9*3, respectively. Celecoxib 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 11337938-2 2001 When compared to cDNA-expressed wild-type CYP2C9 (CYP2C9*1), the Vmax/Km ratio for celecoxib methyl hydroxylation was reduced by 34% and 90% in the presence of recombinant CYP2C9*2 and CYP2C9*3, respectively. Celecoxib 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-58 11337938-2 2001 When compared to cDNA-expressed wild-type CYP2C9 (CYP2C9*1), the Vmax/Km ratio for celecoxib methyl hydroxylation was reduced by 34% and 90% in the presence of recombinant CYP2C9*2 and CYP2C9*3, respectively. Celecoxib 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 11337938-2 2001 When compared to cDNA-expressed wild-type CYP2C9 (CYP2C9*1), the Vmax/Km ratio for celecoxib methyl hydroxylation was reduced by 34% and 90% in the presence of recombinant CYP2C9*2 and CYP2C9*3, respectively. Celecoxib 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 11337938-6 2001 Based on simulations of the in-vitro data obtained with the recombinant CYP2C9 proteins, it was anticipated that the pharmacokinetics of celecoxib (as a much as a five-fold increase in plasma AUC) would be altered (versus CYP2C9*1/*1 subjects) in subjects genotyped heterozygous or homozygous for the CYP2C9*2 (Cys144) or CYP2C9*3 (Leu359) allele. Celecoxib 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 11337938-6 2001 Based on simulations of the in-vitro data obtained with the recombinant CYP2C9 proteins, it was anticipated that the pharmacokinetics of celecoxib (as a much as a five-fold increase in plasma AUC) would be altered (versus CYP2C9*1/*1 subjects) in subjects genotyped heterozygous or homozygous for the CYP2C9*2 (Cys144) or CYP2C9*3 (Leu359) allele. Celecoxib 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 11337938-6 2001 Based on simulations of the in-vitro data obtained with the recombinant CYP2C9 proteins, it was anticipated that the pharmacokinetics of celecoxib (as a much as a five-fold increase in plasma AUC) would be altered (versus CYP2C9*1/*1 subjects) in subjects genotyped heterozygous or homozygous for the CYP2C9*2 (Cys144) or CYP2C9*3 (Leu359) allele. Celecoxib 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 11337938-6 2001 Based on simulations of the in-vitro data obtained with the recombinant CYP2C9 proteins, it was anticipated that the pharmacokinetics of celecoxib (as a much as a five-fold increase in plasma AUC) would be altered (versus CYP2C9*1/*1 subjects) in subjects genotyped heterozygous or homozygous for the CYP2C9*2 (Cys144) or CYP2C9*3 (Leu359) allele. Celecoxib 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 11337938-7 2001 In a subsequent clinical study, the AUC of celecoxib was increased (versus CYP2C9*1/*1 subjects) approximately 2.2-fold (range, 1.6-3-fold) in two CYP2C9*1/*3 subjects and one CYP2C9*3/*3 subject receiving a single oral dose (200 mg) of the drug. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 11337938-7 2001 In a subsequent clinical study, the AUC of celecoxib was increased (versus CYP2C9*1/*1 subjects) approximately 2.2-fold (range, 1.6-3-fold) in two CYP2C9*1/*3 subjects and one CYP2C9*3/*3 subject receiving a single oral dose (200 mg) of the drug. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 11337938-7 2001 In a subsequent clinical study, the AUC of celecoxib was increased (versus CYP2C9*1/*1 subjects) approximately 2.2-fold (range, 1.6-3-fold) in two CYP2C9*1/*3 subjects and one CYP2C9*3/*3 subject receiving a single oral dose (200 mg) of the drug. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 11298070-11 2001 Sildenafil metabolism was inhibited by potent CYP3A4 inhibitors which are used clinically, but was found to be only a weak inhibitor of drug metabolizing enzymes itself, the strongest inhibition occurring against CYP2C9 (Ki = 80 microM). Sildenafil Citrate 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 213-219 11298070-12 2001 CONCLUSIONS: Evidence is provided for CYP3A4 and to a lesser extent CYP2C9-mediated metabolism of sildenafil. Sildenafil Citrate 98-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11298070-13 2001 There is the possibility that elevated plasma concentrations of sildenafil could occur with coadministration of known inhibitors of CYP2C9 or CYP3A4. Sildenafil Citrate 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 71-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Phenobarbital 86-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Rifampin 192-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-6 2001 The concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to rifampicin and phenobarbital paralleled that of CYP3A4 and CYP2B6, the maximum accumulation being reached with 10 microM rifampicin and 100 microM phenobarbital. Phenobarbital 218-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11181490-7 2001 In contrast, dexamethasone produced maximum induction of CYP2C8 and CYP2C9 mRNAs at 0.1 microM while in these conditions neither CYP3A4 nor CYP2B6 was significantly induced. Dexamethasone 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 11181490-8 2001 Moreover, the concentration dependence of CYP2C8 and CYP2C9 mRNAs in response to dexamethasone paralleled that of tyrosine aminotransferase. Dexamethasone 81-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Dexamethasone 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Rifampin 193-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 11181490-9 2001 Furthermore, dexamethasone, which has been recently shown to up-regulate PXR and CAR expression through the glucocorticoid receptor, potentiated CYP2C8 and CYP2C9 mRNA induction in response to rifampicin and phenobarbital. Phenobarbital 208-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 11465391-11 2001 The specificity of inhibition by 8-phenyltheophylline was assessed by measuring its effect on debrisoquine 4-hydroxylase (CYP2D6), terfenadine hydroxylase (CYP3A4), chlorzoxazone 6-hydroxylase (CYP2E1) and tolbutamide 4-hydroxylase (CYP2C9) activities in human liver microsomal fraction. 8-phenyltheophylline 33-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 11181505-9 2001 The metabolic activities of CYP2C9 and CYP2C19 were confirmed by inhibition by sulfaphenazole for CYP2C9 and ticlopidine for CYP2C19. Sulfaphenazole 79-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11181505-9 2001 The metabolic activities of CYP2C9 and CYP2C19 were confirmed by inhibition by sulfaphenazole for CYP2C9 and ticlopidine for CYP2C19. Sulfaphenazole 79-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 11181505-9 2001 The metabolic activities of CYP2C9 and CYP2C19 were confirmed by inhibition by sulfaphenazole for CYP2C9 and ticlopidine for CYP2C19. Ticlopidine 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11307788-5 2001 Gene typing of cytochrome P450 CYP2C9, a liver enzyme responsible for warfarin metabolism, showed that the patient was a carrier of both the mutant alleles (CYP2C9*2/*3) of this enzyme. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 11307788-5 2001 Gene typing of cytochrome P450 CYP2C9, a liver enzyme responsible for warfarin metabolism, showed that the patient was a carrier of both the mutant alleles (CYP2C9*2/*3) of this enzyme. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 11231472-0 2001 Mutations of the CYP2C9 gene and the response to warfarin. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 11207028-0 2001 Inhibition of cytochrome P450 2C9 activity in vitro by 5-hydroxytryptamine and adrenaline. Serotonin 55-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-33 11329260-0 2001 Identification of human CYP2C19 residues that confer S-mephenytoin 4"-hydroxylation activity to CYP2C9. Mephenytoin 53-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 11234294-3 2001 METHOD: In 60 patients on long-term phenytoin therapy the concentration of phenytoin in serum was measured and CYP2C9 genotyping was performed (mutant alleles of CYP2C9 are associated with impaired phenytoin metabolism). Phenytoin 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11234294-3 2001 METHOD: In 60 patients on long-term phenytoin therapy the concentration of phenytoin in serum was measured and CYP2C9 genotyping was performed (mutant alleles of CYP2C9 are associated with impaired phenytoin metabolism). Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11234294-3 2001 METHOD: In 60 patients on long-term phenytoin therapy the concentration of phenytoin in serum was measured and CYP2C9 genotyping was performed (mutant alleles of CYP2C9 are associated with impaired phenytoin metabolism). Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 11234294-5 2001 CYP2C9 genotype and comedication were connected to phenytoin daily dose requirement. Phenytoin 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11741520-5 2001 RESULT: CYP1A2, CYP3A4, and CYP2C9 catalyze the estradiol 2-hydroxylation. Estradiol 48-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 11741520-6 2001 CYP2C9, CYP2C19, and CYP2C8 have high activity in catalyzing 17beta-hydroxy dehydrogenation in cDNA expressed P450, but CYP1A2 is the most important enzyme in catalyzing estradiol 2-hydroxylation. 17beta 61-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11741520-6 2001 CYP2C9, CYP2C19, and CYP2C8 have high activity in catalyzing 17beta-hydroxy dehydrogenation in cDNA expressed P450, but CYP1A2 is the most important enzyme in catalyzing estradiol 2-hydroxylation. Estradiol 170-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11159803-4 2001 DMSO was found to inhibit CYP2C9 and CYP2C19, CYP2E1, and CYP3A4 in a concentration-dependent manner. Dimethyl Sulfoxide 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 11159803-5 2001 At 2% DMSO, the activities for the four isoforms were approximately 40% (CYP2C9), 23% (CYP2C19), and 11% (CYP2E1) of that observed for 0.1% acetonitrile and 45% (CYP3A4) of that observed for 1% acetonitrile. Dimethyl Sulfoxide 6-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 11159803-5 2001 At 2% DMSO, the activities for the four isoforms were approximately 40% (CYP2C9), 23% (CYP2C19), and 11% (CYP2E1) of that observed for 0.1% acetonitrile and 45% (CYP3A4) of that observed for 1% acetonitrile. acetonitrile 140-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 11159803-5 2001 At 2% DMSO, the activities for the four isoforms were approximately 40% (CYP2C9), 23% (CYP2C19), and 11% (CYP2E1) of that observed for 0.1% acetonitrile and 45% (CYP3A4) of that observed for 1% acetonitrile. acetonitrile 194-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 11159803-7 2001 Methanol was found to inhibit CYP2C9 and CYP2E1 activities, but to a lesser extent than DMSO. Methanol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 11207028-1 2001 In the present study, the occurrence of a modulatory effect of 14 neurotransmitters, precursors and metabolites on the cytochrome P450 2C9 (CYP2C9) enzyme activity, as determined by diclofenac 4-hydroxylation, was studied in human liver microsomes. Diclofenac 182-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 11213860-0 2001 Implications of cytochrome P450 2C9 polymorphism on warfarin metabolism and dosing. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-35 11213860-3 2001 S-Warfarin, the more potent isomer, is metabolized primarily by CYP2C9. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 11213860-6 2001 Clinical studies showed an increased plasma level of S-warfarin, decreased clearance of S-warfarin, increased frequency of bleeding, and prolongation of hospitalization in patients with variant CYP2C9 alleles. Warfarin 53-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 11213860-6 2001 Clinical studies showed an increased plasma level of S-warfarin, decreased clearance of S-warfarin, increased frequency of bleeding, and prolongation of hospitalization in patients with variant CYP2C9 alleles. Sulfur 53-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 11213860-6 2001 Clinical studies showed an increased plasma level of S-warfarin, decreased clearance of S-warfarin, increased frequency of bleeding, and prolongation of hospitalization in patients with variant CYP2C9 alleles. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 11206048-4 2001 The genotypes of CYP2C9 and 2C19, the enzymes responsible for phenytoin metabolism, were homozygous for the wild-type alleles (CYP2C9*1/*1 and 2C19*1/ *1). Phenytoin 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 11206048-4 2001 The genotypes of CYP2C9 and 2C19, the enzymes responsible for phenytoin metabolism, were homozygous for the wild-type alleles (CYP2C9*1/*1 and 2C19*1/ *1). Phenytoin 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 11206048-5 2001 The interaction may be a result of inhibition of both CYP2C9 and 2C19 by fluvoxamine. Fluvoxamine 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 11207028-0 2001 Inhibition of cytochrome P450 2C9 activity in vitro by 5-hydroxytryptamine and adrenaline. Epinephrine 79-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-33 11207028-1 2001 In the present study, the occurrence of a modulatory effect of 14 neurotransmitters, precursors and metabolites on the cytochrome P450 2C9 (CYP2C9) enzyme activity, as determined by diclofenac 4-hydroxylation, was studied in human liver microsomes. Diclofenac 182-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-138 11139472-4 2001 The selective CYP 2C9 inhibitor sulfaphenazole and the superoxide anion (O(2-)) scavengers Tiron and nordihydroguaretic acid also induced a leftward shift in the NO-mediated concentration-relaxation curve to bradykinin. Sulfaphenazole 32-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-21 11139472-4 2001 The selective CYP 2C9 inhibitor sulfaphenazole and the superoxide anion (O(2-)) scavengers Tiron and nordihydroguaretic acid also induced a leftward shift in the NO-mediated concentration-relaxation curve to bradykinin. 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt 91-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-21 11139472-0 2001 Endothelium-derived hyperpolarizing factor synthase (Cytochrome P450 2C9) is a functionally significant source of reactive oxygen species in coronary arteries. Reactive Oxygen Species 114-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 11139472-4 2001 The selective CYP 2C9 inhibitor sulfaphenazole and the superoxide anion (O(2-)) scavengers Tiron and nordihydroguaretic acid also induced a leftward shift in the NO-mediated concentration-relaxation curve to bradykinin. Masoprocol 101-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-21 11139472-5 2001 CYP activity and O(2-) production, determined in microsomes prepared from cells overexpressing CYP 2C9, were almost completely inhibited by sulfaphenazole. Superoxides 17-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-102 11139472-5 2001 CYP activity and O(2-) production, determined in microsomes prepared from cells overexpressing CYP 2C9, were almost completely inhibited by sulfaphenazole. Sulfaphenazole 140-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-102 11139472-11 2001 These results suggest that a CYP isozyme homologous to CYP 2C9 is a physiologically relevant generator of ROS in coronary endothelial cells and modulates both vascular tone and homeostasis. Reactive Oxygen Species 106-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-62 11730569-0 2001 Effects of CYP2C19 genotype and CYP2C9 on fluoxetine N-demethylation in human liver microsomes. Fluoxetine 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 11475198-9 2001 Interactions associated with CYP2C9-interference may, however, be present for fluvastatin. Fluvastatin 78-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 11730569-10 2001 CONCLUSION: Cytochrome P-450 CYP2C9 is likely to be a major CYP isoform catalyzing fluoxetine N-demethylation in human liver microsomes at a substrate concentration close to the therapeutic level, while polymorphic CYP2C19 may play a more important role in this metabolic pathway at high substrate concentration. fluoxetine n 83-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 11180037-0 2001 Fluvoxamine inhibits the CYP2C9 catalyzed biotransformation of tolbutamide. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11135730-8 2001 All of the CYPs evaluated were capable of hydroxylating propofol; however, CYP2B6 and CYP2C9 were most active. Propofol 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 11135730-11 2001 CONCLUSIONS: Cytochrome P-450 2B6, and to a lesser extent CYP2C9, contribute to the oxidative metabolism of propofol. Propofol 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 11180037-0 2001 Fluvoxamine inhibits the CYP2C9 catalyzed biotransformation of tolbutamide. Tolbutamide 63-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11180037-1 2001 OBJECTIVE: Our objective was to examine the interaction between fluvoxamine and tolbutamide to confirm that fluvoxamine inhibits CYP2C9. Tolbutamide 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 11180037-1 2001 OBJECTIVE: Our objective was to examine the interaction between fluvoxamine and tolbutamide to confirm that fluvoxamine inhibits CYP2C9. Fluvoxamine 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 11368292-5 2001 The 6-hydroxy and N-desisopropyl fluvastatin metabolites are exclusively generated by cytochrome P450 (CYP) 2C9 and do not accumulate in the blood. 6-hydroxy and n-desisopropyl fluvastatin 4-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-111 11180037-13 2001 CONCLUSION: Fluvoxamine is a moderate inhibitor of CYP2C9 in vivo. Fluvoxamine 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 11735605-5 2001 On the other hand, rifampicin is a nonspecific inducer of CYP-mediated metabolism (especially of CYP2C9, CYP2C19 and CYP3A4) and may therefore cause therapeutic failure of other coadministered drugs by increasing their clearance. Rifampin 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 11368292-6 2001 CYP2C9, CYP3A4, CYP2C8 and CYP2D6 form the 5-hydroxy fluvastatin metabolite. 5-hydroxy fluvastatin 43-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11368292-13 2001 In addition to being a CYP2C9 substrate, fluvastatin demonstrates inhibitory effects on this isoenzyme in vitro and in vivo. Fluvastatin 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 11368292-14 2001 In human liver microsomes, fluvastatin significantly inhibits the hydroxylation of 2 CYP2C9 substrates, tolbutamide and diclofenac. Fluvastatin 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Diclofenac 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Tolbutamide 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11523725-12 2001 Human liver microsomes obtained from individuals heterozygous for CYP2C9*3 showed significantly reduced (S)-warfarin 7-hydroxylation as compared with those obtained from individuals genotyped as CYP2C9*1. Warfarin 104-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 11523725-13 2001 The influence of the CYP2C9*3 allele on the in vivo pharmacokinetics of (S)-warfarin has been studied in Japanese patients. Warfarin 72-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 11523725-14 2001 Patients with the homozygous CYP2C9*3 genotype, as well as those with the heterozygous CYP2C9*1/*3 genotype, had significantly reduced clearance of (S)-warfarin (by 90 and 60%, respectively) compared with those with homozygous CYP2C9*1. Warfarin 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 11523725-14 2001 Patients with the homozygous CYP2C9*3 genotype, as well as those with the heterozygous CYP2C9*1/*3 genotype, had significantly reduced clearance of (S)-warfarin (by 90 and 60%, respectively) compared with those with homozygous CYP2C9*1. Warfarin 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 11523725-14 2001 Patients with the homozygous CYP2C9*3 genotype, as well as those with the heterozygous CYP2C9*1/*3 genotype, had significantly reduced clearance of (S)-warfarin (by 90 and 60%, respectively) compared with those with homozygous CYP2C9*1. Warfarin 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 11523725-15 2001 The maintenance dosages of warfarin required in Japanese patients with heterozygous and homozygous CYP2C9*3 mutations were significantly lower than those in patients with CYP2C9*1/*1. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 11523725-16 2001 In addition, 86% of British patients exhibiting adequate therapeutic responses with lower maintenance dosages of warfarin (<1.5 mg/day) had either the CYP2C9*2 or CYP2C9*3 mutation singly or in combination, whereas only 38% of randomly selected patients receiving warfarin carried the corresponding mutations. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Glyburide 70-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Glyburide 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Losartan 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11368292-15 2001 The oral clearances of the CYP2C9 substrates diclofenac, tolbutamide, glibenclamide (glyburide) and losartan are reduced by 15 to 25% when coadministered with fluvastatin. Fluvastatin 159-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11523725-18 2001 These data indicate that the CYP2C9*3 allele may be associated with retarded elimination of (S)-warfarin and the resulting clinical effects. Warfarin 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 11523726-2 2001 Although irbesartan does not require biotransformation for its pharmacological activity, it does undergo metabolism via the cytochrome P450 (CYP) 2C9 isoenzyme and negligible metabolism by the CYP3A4 isoenzyme. Irbesartan 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-149 11124226-5 2001 Niclosamide had substrate-dependent contrasting effects on CYP2C9 activity with an apparent activation (400%) of 7-methoxy-4-trifluoromethylcoumarin demethylase activity and potent inhibition (K(i) = 6.00 microM) of diclofenac 4-hydroxylase activity. Niclosamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 11124228-0 2001 Interaction of delavirdine with human liver microsomal cytochrome P450: inhibition of CYP2C9, CYP2C19, and CYP2D6. Delavirdine 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 11124228-3 2001 The interaction of delavirdine with CYP2C9 was examined with pooled human liver microsomes using diclofenac 4"-hydroxylation as a reporter of CYP2C9 catalytic activity. Delavirdine 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 11124228-3 2001 The interaction of delavirdine with CYP2C9 was examined with pooled human liver microsomes using diclofenac 4"-hydroxylation as a reporter of CYP2C9 catalytic activity. Delavirdine 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 11124228-8 2001 Several analogs of delavirdine showed similar inhibition of CYP2C9. Delavirdine 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 11124228-12 2001 These results, along with previously reported experiments, indicate that delavirdine can partially inhibit CYP2C9, -2C19, -2D6, and -3A4, although the degree of inhibition in vivo would be subject to a variety of additional factors. Delavirdine 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 11887965-3 2001 Differential response to phenytoin, for example, is related to interindividual genetic differences in the metabolic enzyme CYP2C9, and to a lesser extent, CYP2C19. Phenytoin 25-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 11294368-0 2001 Is diclofenac a valuable CYP2C9 probe in humans? diclofenac a 3-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11294368-2 2001 The in vitro CYP2C9-specific substrate diclofenac might be a valuable, well-tolerated probe candidate. Diclofenac 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 11294368-3 2001 In order to validate diclofenac as an in vivo CYP2C9 probe, we planned to show that urinary 4"-hydroxydiclofenac/diclofenac metabolic ratio (MR) would correlate to the apparent partial metabolic clearance of diclofenac into 4"-hydroxydiclofenac (Clmet). Diclofenac 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 11294368-10 2001 However, since we found a lower metabolic clearance of diclofenac in heterozygous CYP2C9 subjects, as observed with other CYP2C9 substrates, diclofenac, in another galenic form, might be a potential probe to quantify CYP2C9 activity in humans. Diclofenac 55-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 11294368-10 2001 However, since we found a lower metabolic clearance of diclofenac in heterozygous CYP2C9 subjects, as observed with other CYP2C9 substrates, diclofenac, in another galenic form, might be a potential probe to quantify CYP2C9 activity in humans. Diclofenac 55-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 11294368-10 2001 However, since we found a lower metabolic clearance of diclofenac in heterozygous CYP2C9 subjects, as observed with other CYP2C9 substrates, diclofenac, in another galenic form, might be a potential probe to quantify CYP2C9 activity in humans. Diclofenac 55-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 11294368-10 2001 However, since we found a lower metabolic clearance of diclofenac in heterozygous CYP2C9 subjects, as observed with other CYP2C9 substrates, diclofenac, in another galenic form, might be a potential probe to quantify CYP2C9 activity in humans. Diclofenac 141-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 11908757-0 2001 The predictive value of MDR1, CYP2C9, and CYP2C19 polymorphisms for phenytoin plasma levels. Phenytoin 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 11908757-8 2001 A combined analysis of variable alleles of CYP2C9, 2C19 and MDR1 revealed that the number of mutant CYP2C9 alleles is a major determinant, the number of MDR1*T alleles further contributes to the prediction of phenytoin plasma levels and CYP2C19*2 does not explain individual variability. Phenytoin 209-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11334262-2 2001 The inhibitory effects of cimetidine, nizatidine and omeprazole on the metabolic activity of CYP2C9, 2C19, 2D6 and 3A were investigated in human liver microsomes. Cimetidine 26-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 11334262-2 2001 The inhibitory effects of cimetidine, nizatidine and omeprazole on the metabolic activity of CYP2C9, 2C19, 2D6 and 3A were investigated in human liver microsomes. Nizatidine 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 12579866-7 2001 CONCLUSION: There were significant individual differences in the pharmacokinetics of meloxicam in Chinese volunteers, which may be due to the genetic polymorphism of CYP2C9. Meloxicam 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 11908757-8 2001 A combined analysis of variable alleles of CYP2C9, 2C19 and MDR1 revealed that the number of mutant CYP2C9 alleles is a major determinant, the number of MDR1*T alleles further contributes to the prediction of phenytoin plasma levels and CYP2C19*2 does not explain individual variability. Phenytoin 209-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 11334262-2 2001 The inhibitory effects of cimetidine, nizatidine and omeprazole on the metabolic activity of CYP2C9, 2C19, 2D6 and 3A were investigated in human liver microsomes. Omeprazole 53-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 11908757-9 2001 The regression equation that fitted the data best included the number of mutant CYP2C9 and MDR*T alleles as predictory variables and explained 15.4% of the variability of phenytoin data (r2 = 0.154, P = 0.0002). Phenytoin 171-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 11908757-10 2001 Furthermore, analysis of CYP2C9 and MDR1 genotypes in 35 phenytoin-treated patients recruited from therapeutic drug monitoring showed that combined CYP2C9 and MDR1 analysis has some predictive value not only in the controlled settings of a clinical trial, but also in the daily clinical practice. Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 11908757-10 2001 Furthermore, analysis of CYP2C9 and MDR1 genotypes in 35 phenytoin-treated patients recruited from therapeutic drug monitoring showed that combined CYP2C9 and MDR1 analysis has some predictive value not only in the controlled settings of a clinical trial, but also in the daily clinical practice. Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 11908770-0 2001 CYP2C9*3 influences the metabolism and the drug-interaction of candesartan in vitro. candesartan 63-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11908770-1 2001 Candesartan cilexetil is an angiotensin II receptor antagonist, and candesartan, its active metabolite, is metabolized by CYP2C9. candesartan 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 11908770-3 2001 We characterized the kinetics of candesartan by CYP2C9*1/*1 and CYP2C9*1/*3 in human liver microsomes. candesartan 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 11908770-3 2001 We characterized the kinetics of candesartan by CYP2C9*1/*1 and CYP2C9*1/*3 in human liver microsomes. candesartan 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 11908770-5 2001 Subsequently, CYP2C9*1 and CYP2C9*3 (Leu359) were expressed in yeast, and the kinetics of candesartan were determined. candesartan 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 11908770-5 2001 Subsequently, CYP2C9*1 and CYP2C9*3 (Leu359) were expressed in yeast, and the kinetics of candesartan were determined. candesartan 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. Rifampin 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. glimepiride 66-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. Rifampin 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11136298-7 2000 CONCLUSIONS: The effects of rifampicin on the pharmacokinetics of glimepiride suggest that rifampicin induced the CYP2C9-mediated metabolism of glimepiride and thereby slightly increased its systemic clearance. glimepiride 144-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 11095582-6 2000 The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K"(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation. furafylline 145-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 661-667 11095582-6 2000 The involvement of CYP1A2 in DMXAA metabolism by human livers was demonstrated by the following: 1) the potent inhibition of DMXAA metabolism by furafylline (k(inact) = 0.23 +/- 0.04 min(-1), K"(app) = 15.6 +/- 6.7 microM) and alpha-naphthoflavone (K(i) = 0.036 microM), but not by cimetidine, ketoconazole, tolbutamide, quinidine, chlorzoxazone, diethyldithiocarbamate, troleandomycin, and sulfaphenazole; 2) when incubated with human lymphoblastoid cell microsomes containing cDNA-expressed CYP isoenzymes, DMXAA was metabolized only by CYP1A2, with an apparent K(m) of 6.2 +/- 1.5 microM and V(max) of 0.014 +/- 0.001 nmol/min/mg, but not by CYP2A6, CYP2B6, CYP2C9 (Arg(144)), CYP2C19, CYP2D6 (Val(374)), CYP2E1, and CYP3A4; 3) a significant correlation (r = 0.90; P <.001) between 6-methylhydroxylation of DMXAA and 7-ethoxyresorufin O-deethylation; and 4) a significant correlation (r = 0.75; P <.01) between the CYP1A protein level determined by Western blots and DMXAA 6-methylhydroxylation. vadimezan 29-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 661-667 11129071-8 2000 This method provides a sensitive and specific assay for the detection of flurbiprofen and 4"-hydoxyflurbiprofen in urine and plasma and is suitable for use in in vivo studies evaluating the regulation of CYP2C9 activity. Flurbiprofen 73-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 204-210 11038165-0 2000 Formation of a dihydroxy metabolite of phenytoin in human liver microsomes/cytosol: roles of cytochromes P450 2C9, 2C19, and 3A4. Phenytoin 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-119 11038154-4 2000 Of various cDNA-expressed human CYPs, only CYP2C9 hydroxylated (S)-acenocoumarol. Acenocoumarol 63-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). 3",4"-dihpph 88-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 11038154-6 2000 CYP2C9 also mediated the 6-, 7-, and 8-hydroxylations of (R)-acenocoumarol with K(m) values three to four times and V(max) values one-sixth times those of (S)-acenocoumarol. (R)-Acenocoumarol 57-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11038161-7 2000 Tolbutamide was metabolized by CYP2C9 (70%) and CYP2C19 (30%), diazepam by CYP2C19 (100%), ibuprofen by CYP2C9 (90%) and CYP2C19 (10%), and omeprazole by CYP2C19 (68%) and CYP3A4 (32%). Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 11038161-7 2000 Tolbutamide was metabolized by CYP2C9 (70%) and CYP2C19 (30%), diazepam by CYP2C19 (100%), ibuprofen by CYP2C9 (90%) and CYP2C19 (10%), and omeprazole by CYP2C19 (68%) and CYP3A4 (32%). Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11038154-6 2000 CYP2C9 also mediated the 6-, 7-, and 8-hydroxylations of (R)-acenocoumarol with K(m) values three to four times and V(max) values one-sixth times those of (S)-acenocoumarol. Acenocoumarol 155-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). 3"-hydroxy-dph 154-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 11038154-14 2000 The results demonstrate that (S)-acenocoumarol is hydroxylated by a single enzyme, namely CYP2C9. Acenocoumarol 29-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 11038154-15 2000 CYP2C9 is also the main enzyme in the 7-hydroxylation of (R)-acenocoumarol. (R)-Acenocoumarol 57-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 11038161-7 2000 Tolbutamide was metabolized by CYP2C9 (70%) and CYP2C19 (30%), diazepam by CYP2C19 (100%), ibuprofen by CYP2C9 (90%) and CYP2C19 (10%), and omeprazole by CYP2C19 (68%) and CYP3A4 (32%). Ibuprofen 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 11038165-7 2000 Of 10 cDNA-expressed human P450 enzymes examined, CYP2C19, CYP2C9, and CYP3A4 catalyzed 3",4"-diHPPH formation from the primary hydroxylated metabolites (3"-hydroxy-DPH and 4"-HPPH). hydroxyphenytoin 173-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 11038165-9 2000 These results suggest that CYP2C9, CYP2C19, and CYP3A4 all have catalytic activities in 3",4"-diHPPH formation from primary hydroxylated metabolites in human liver and that the hepatic contents of these three P450 forms determine which P450 enzymes play major roles of DPH oxidation in individual humans. 3",4"-dihpph 88-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11038165-9 2000 These results suggest that CYP2C9, CYP2C19, and CYP3A4 all have catalytic activities in 3",4"-diHPPH formation from primary hydroxylated metabolites in human liver and that the hepatic contents of these three P450 forms determine which P450 enzymes play major roles of DPH oxidation in individual humans. Phenytoin 269-272 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 11186138-0 2000 The possession of the CYP2C9*3 allele is associated with low dose requirement of acenocoumarol. Acenocoumarol 81-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 10991840-0 2000 Cytochrome P-450 2C9 sensitizes human prostate tumor cells to cyclophosphamide via a bystander effect. Cyclophosphamide 62-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-20 11127854-1 2000 Cytochrome P450 CYP2C9 gene variants have been associated with hyperresponsiveness to small doses of warfarin and a higher bleeding complication rate. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 11127854-4 2000 The adjusted dose required of warfarin was higher among patients with the CYP2C9*1 haplotype (5.6 mg) than those of patients carrying the CYP2C9*2 (4.7 mg; p = 0.007, Scheffe"s test) or the CYP2C9*3 haplotype (4.0 mg; p <0.001, Scheffe"s test). Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 11127854-4 2000 The adjusted dose required of warfarin was higher among patients with the CYP2C9*1 haplotype (5.6 mg) than those of patients carrying the CYP2C9*2 (4.7 mg; p = 0.007, Scheffe"s test) or the CYP2C9*3 haplotype (4.0 mg; p <0.001, Scheffe"s test). Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11127854-4 2000 The adjusted dose required of warfarin was higher among patients with the CYP2C9*1 haplotype (5.6 mg) than those of patients carrying the CYP2C9*2 (4.7 mg; p = 0.007, Scheffe"s test) or the CYP2C9*3 haplotype (4.0 mg; p <0.001, Scheffe"s test). Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11127854-8 2000 CYP2C9 gene variants modulate the anticoagulant effect of the dose of warfarin prescribed. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10991840-5 2000 Cytotoxicity was studied by exposing the cells to 0.01 to 4 mM CPA in the presence or absence of sulfaphenazole, a CYP2C9-specific inhibitor. Sulfaphenazole 97-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 10991840-7 2000 H2C9 cells quickly metabolized diclofenac, indicating the presence of high levels of CYP2C9. Diclofenac 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 10991840-12 2000 H2C9 cells mediated a bystander killing effect for CYP2C9-negative PPC-1 cells, reducing the IC(50) of CPA from about 14 to 3.62+/-0.73 mM in PPC-1 cells when they were cocultured with H2C9 cells. Cyclophosphamide 103-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10991840-1 2000 The goal of the present study was to examine the ability of cytochrome P450-2C9 (CYP2C9) to activate cyclophosphamide (CPA) and elicit tumor cell death. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-79 10991840-1 2000 The goal of the present study was to examine the ability of cytochrome P450-2C9 (CYP2C9) to activate cyclophosphamide (CPA) and elicit tumor cell death. Cyclophosphamide 101-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10991840-1 2000 The goal of the present study was to examine the ability of cytochrome P450-2C9 (CYP2C9) to activate cyclophosphamide (CPA) and elicit tumor cell death. Cyclophosphamide 119-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-79 10991840-1 2000 The goal of the present study was to examine the ability of cytochrome P450-2C9 (CYP2C9) to activate cyclophosphamide (CPA) and elicit tumor cell death. Cyclophosphamide 119-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10991840-3 2000 The catalytic activity present in cell lines was examined by measuring the conversion of diclofenac, a CYP2C9-specific substrate, to its 4"-hydroxy metabolite by high-pressure liquid chromatography. Diclofenac 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 11317178-3 2000 The clinical significance of this event as well as a possible interaction between clopidogrel and other drugs metabolized by the cytochrome P450 2C9 pathways coadministered to this patient are discussed. Clopidogrel 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-148 10997938-6 2000 Scaling the intrinsic clearance values for the individual CYP enzymes to estimate contributions of each in human liver microsomes suggested that CYP2D6, CYP2C9, and CYP3A4 contribute the greatest amount of fluoxetine N-demethylation in human liver microsomes. Fluoxetine 206-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 10997938-7 2000 These data were corroborated with the examination of the effects of CYP-specific inhibitors quinidine (CYP2D6), sulfaphenazole (CYP2C9), and ketoconazole (CYP3A4) on fluoxetine N-demethylation in pooled human liver microsomes. Sulfaphenazole 112-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 10997938-7 2000 These data were corroborated with the examination of the effects of CYP-specific inhibitors quinidine (CYP2D6), sulfaphenazole (CYP2C9), and ketoconazole (CYP3A4) on fluoxetine N-demethylation in pooled human liver microsomes. Fluoxetine 166-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 11315104-10 2000 Using human beta-lymphoblastoid cell microsomes containing cDNA-expressed CYP isoforms, 20 microM BFC was metabolized by CYP1A2 and CYP3A4, with lower rates of metabolism being observed with CYP2C9 and CYP2C19. 7-benzyloxy-4-trifluoromethylcoumarin 98-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 11315104-19 2000 BFC metabolism appears to be primarily catalysed by CYP1A2 and CYP3A4, with possibly some contribution by CYP2C9, CYP2C19 and perhaps other CYP isoforms. 7-benzyloxy-4-trifluoromethylcoumarin 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 10961881-0 2000 Influence of cytochrome P-450 CYP2C9 polymorphisms on warfarin sensitivity and risk of over-anticoagulation in patients on long-term treatment. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10961881-1 2000 Cytochrome P-450 2C9 is the principle enzyme that terminates the anticoagulant effect of warfarin. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-20 10961881-11 2000 This study confirmed an association between CYP2C9 genotype and warfarin sensitivity. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 10960410-10 2000 CYP2C9 showed a significantly rapid metabolism of R(+)-enantiomer, suggesting that CYP2C9 is mainly involved in the enantioselective metabolism of thiamylal. Thiamylal 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10960410-10 2000 CYP2C9 showed a significantly rapid metabolism of R(+)-enantiomer, suggesting that CYP2C9 is mainly involved in the enantioselective metabolism of thiamylal. Thiamylal 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 10955816-9 2000 Although sulfaphenazole (an inhibitor of CYP2C9) and S-mephenytoin (an inhibitor of CYP2C19) partially inhibited the disappearance of MPA, no effect of the anti-CYP2C antibody was observed. Sulfaphenazole 9-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 11026737-4 2000 At 10 microM PER, a concentration consistent with anticipated in vivo liver concentrations, CYP3A4 and CYP2C9 contributed 50% and 35%, respectively, to PER-N-demethylation. Nitrogen 156-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 11026737-14 2000 CONCLUSIONS: Alterations in the activity of CYP3A4, CYP2C9 and FMO3 through genetic polymorphisms, enzyme induction or inhibition bear the potential to cause clinically significant changes in perazine clearance. Perazine 192-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Rifampin 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10924340-4 2000 In parallel, IL-6 decreases both rifampicin- and phenobarbital-mediated induction of CYP2B6, CYP2C8, CYP2C9, and CYP3A4. Phenobarbital 49-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10930961-15 2000 The more pronounced effect of ritonavir may be attributed to its additional potent inhibition of CYP2C9. Ritonavir 30-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 11029845-8 2000 Lovastatin, simvastatin, and atorvastatin are substrates of CYP3A4, whereas fluvastatin is metabolized by CYP2C9. Fluvastatin 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 11029845-10 2000 Compared with other statins, pravastatin thus has a reduced potential for drug interactions with other substrates, inhibitors, or inducers of the CYP3A4 and CYP2C9 systems. Pravastatin 29-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 10901692-7 2000 In contrast, S-mephenytoin inhibited DDS-NHY by CYP2C9, CYP2C18, and CYP2C19 by 27 +/- 2, 49 +/- 1, and 32 +/- 4%, respectively. Mephenytoin 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 10976551-3 2000 We studied the possible correlation of the formation clearance of 3-hydroxyquinidine with probe-based assays for CYP1A2, CYP2C9, CYP2C19, and CYP2D6. 3-hydroxyquinidine 66-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 10901692-2 2000 Sulfaphenazole (2.16 microM) and tolbutamide (500 microM), selective inhibitors of CYP2C9 (or 2C8/9), inhibited DDS-NHY by 48 +/- 14 and 41 +/- 15%, respectively. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 10901692-7 2000 In contrast, S-mephenytoin inhibited DDS-NHY by CYP2C9, CYP2C18, and CYP2C19 by 27 +/- 2, 49 +/- 1, and 32 +/- 4%, respectively. Fumigant 93 37-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 10901692-2 2000 Sulfaphenazole (2.16 microM) and tolbutamide (500 microM), selective inhibitors of CYP2C9 (or 2C8/9), inhibited DDS-NHY by 48 +/- 14 and 41 +/- 15%, respectively. Tolbutamide 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 10901707-6 2000 Although the CYP2C9 inhibitor sulfaphenazole (100 microM) inhibited MEGX formation by about 30%, recombinant human CYP2C9 showed very low catalytic activity, suggesting a negligible role for this enzyme in lidocaine N-deethylation. Sulfaphenazole 30-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 10901692-8 2000 Because CYP2C18 and CYP19 are expressed at low concentrations in the human liver, these observations indicate that at clinical DDS concentrations, CYP2C9 is a major and CYP2C8 is a likely minor contributor to DDS-NHY in human liver microsomes. Fumigant 93 127-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10901712-3 2000 The Catalyst models generated from multiple conformers of competitive inhibitors of CYP2C9 activities contained at least one hydrophobic and two hydrogen bond acceptor/donor regions. Hydrogen 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 10901692-2 2000 Sulfaphenazole (2.16 microM) and tolbutamide (500 microM), selective inhibitors of CYP2C9 (or 2C8/9), inhibited DDS-NHY by 48 +/- 14 and 41 +/- 15%, respectively. dds-nhy 112-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 10901692-3 2000 The apparent Michaelis-Menten Km values for DDS-NHY by cloned CYP2C8, CYP2C9, CYP2C18, and CYP2C19 were 75 microM, 31 microM, 25 microM, and greater than 1 mM, respectively. Fumigant 93 44-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 10948089-4 2000 The CYP inducer beta-naphthoflavone and the Ca(2+) antagonist nifedipine significantly increased CYP2C mRNA but did not change the expression of CYP2J or CYP2B. beta-Naphthoflavone 16-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-102 10948089-4 2000 The CYP inducer beta-naphthoflavone and the Ca(2+) antagonist nifedipine significantly increased CYP2C mRNA but did not change the expression of CYP2J or CYP2B. Nifedipine 62-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-102 10948089-5 2000 To determine the relationship between CYP2C expression and EDHF production in native endothelial cells, we incubated porcine coronary arteries with nifedipine. edhf 59-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-43 10948089-6 2000 Nifedipine enhanced endothelial CYP2C protein expression, as well as the generation of 11,12-epoxyeicosatrienoic acid. Nifedipine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-37 10948089-8 2000 The specific CYP2C9 inhibitor sulfaphenazole, on the other hand, significantly attenuated EDHF-mediated hyperpolarization and relaxation. Sulfaphenazole 30-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 10948089-8 2000 The specific CYP2C9 inhibitor sulfaphenazole, on the other hand, significantly attenuated EDHF-mediated hyperpolarization and relaxation. edhf 90-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 10871299-7 2000 The flavonoid compound I3,II8-biapigenin was shown to be a potent, competitive inhibitor of CYP3A4, CYP2C9, and CYP1A2 activities with K(i) values of 0.038, 0.32, and 0.95 microM, respectively. Flavonoids 4-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 10975605-0 2000 Relationship of polymorphism in CYP2C9 to genetic susceptibility to diclofenac-induced hepatitis. Diclofenac 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 10975605-2 2000 Adduct formation may be due to metabolism of diclofenac via an alternative pathway rather than via its major 4"-hydroxylation pathway mediated by the cytochrome P450 CYP2C9. Diclofenac 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 10975605-3 2000 We hypothesized that possession of variant CYP2C9 alleles might be a risk factor for diclofenac-induced hepatotoxicity, since the allelic variants CYP2C9*2 and CYP2C9*3 may be associated with impaired metabolism compared to the wild-type (CYP2C9*1). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 10975605-3 2000 We hypothesized that possession of variant CYP2C9 alleles might be a risk factor for diclofenac-induced hepatotoxicity, since the allelic variants CYP2C9*2 and CYP2C9*3 may be associated with impaired metabolism compared to the wild-type (CYP2C9*1). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10975605-3 2000 We hypothesized that possession of variant CYP2C9 alleles might be a risk factor for diclofenac-induced hepatotoxicity, since the allelic variants CYP2C9*2 and CYP2C9*3 may be associated with impaired metabolism compared to the wild-type (CYP2C9*1). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10975605-3 2000 We hypothesized that possession of variant CYP2C9 alleles might be a risk factor for diclofenac-induced hepatotoxicity, since the allelic variants CYP2C9*2 and CYP2C9*3 may be associated with impaired metabolism compared to the wild-type (CYP2C9*1). Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10975605-7 2000 The relationship between CYP2C9 genotype and susceptibility to diclofenac-induced hepatotoxicity was further examined by genotyping 24 patients with diclofenac-induced hepatotoxicity together with 100 healthy controls for the CYP2C9*2 and CYP2C9*3 alleles. Diclofenac 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 10923005-8 2000 Compared to a wild-type genotype, the presence of the CYP2C9*2, CYP2C9*3, or CYP2A6*2 allele was associated with a significant reduction in weekly warfarin dose (mean weekly warfarin dose [+/- SE] for wild-type genotype was 0.397 +/- 0.024 mg/kg/wk vs 0.307 +/- 0.03 mg/kg/wk for carriers of CYP2C9*2, CYP2C9*3, or CYP2A6*2 polymorphism; P =.03). Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10871299-7 2000 The flavonoid compound I3,II8-biapigenin was shown to be a potent, competitive inhibitor of CYP3A4, CYP2C9, and CYP1A2 activities with K(i) values of 0.038, 0.32, and 0.95 microM, respectively. ii8-biapigenin 26-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 10871299-8 2000 Hyperforin was a potent noncompetitive inhibitor of CYP2D6 activity (K(i) = 1.5 microM) and competitive inhibitor of CYP2C9 and CYP3A4 activities (K(i) = 1.8 and 0.48 microM, respectively). hyperforin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 10820139-9 2000 At high concentrations of modafinil (>/=100 microM), the mean activity of CYP2C9 was decreased (up to 60%) relative to that in the solvent controls. Modafinil 26-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 10820139-10 2000 Overall, modafinil was shown to have effects on human hepatic CYP1A2, CYP2B6, CYP2C9, CYP2C19, and CYP3A4/5 activities in vitro. Modafinil 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 10895987-8 2000 Clear inhibition was found for denitronifedipine oxidation (CYP3A4; IC50=29 microM and 46 microM) and S(-)-warfarin 7-hydroxylation (CYP2C9; IC50=43 microM and 45 microM). Warfarin 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 10952477-8 2000 The mechanism of the increased plasma concentrations and prolonged elimination of fluvastatin is probably inhibition of the CYP2C9-mediated metabolism of fluvastatin by fluconazole. Fluvastatin 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 10952477-8 2000 The mechanism of the increased plasma concentrations and prolonged elimination of fluvastatin is probably inhibition of the CYP2C9-mediated metabolism of fluvastatin by fluconazole. Fluvastatin 154-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 10952477-8 2000 The mechanism of the increased plasma concentrations and prolonged elimination of fluvastatin is probably inhibition of the CYP2C9-mediated metabolism of fluvastatin by fluconazole. Fluconazole 169-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 10952477-9 2000 Care should be taken if fluconazole or other potent inhibitors of CYP2C9 are prescribed to patients using fluvastatin. Fluconazole 24-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10952477-9 2000 Care should be taken if fluconazole or other potent inhibitors of CYP2C9 are prescribed to patients using fluvastatin. Fluvastatin 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10895987-11 2000 With therapeutic silibinin peak plasma concentrations of 0.6 microM and biliary concentrations up to 200 microM, metabolic interactions with xenobiotics metabolised by CYP3A4 or CYP2C9 cannot be excluded. Silybin 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 10850388-0 2000 Catalytic activity of three variants (Ile, Leu, and Thr) at amino acid residue 359 in human CYP2C9 gene and simultaneous detection using single-strand conformation polymorphism analysis. Threonine 52-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 10850388-1 2000 This study evaluated the catalytic activity of three variants (Ile, Leu, and Thr) at codon 359 of CYP2C9 enzymes expressed in a yeast cDNA expression system, and then established single-strand conformation polymorphism (PCR-SSCP) analysis for simultaneous detection as a screening method. Isoleucine 63-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 10850388-0 2000 Catalytic activity of three variants (Ile, Leu, and Thr) at amino acid residue 359 in human CYP2C9 gene and simultaneous detection using single-strand conformation polymorphism analysis. Isoleucine 38-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 10850388-1 2000 This study evaluated the catalytic activity of three variants (Ile, Leu, and Thr) at codon 359 of CYP2C9 enzymes expressed in a yeast cDNA expression system, and then established single-strand conformation polymorphism (PCR-SSCP) analysis for simultaneous detection as a screening method. Leucine 68-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 10850388-0 2000 Catalytic activity of three variants (Ile, Leu, and Thr) at amino acid residue 359 in human CYP2C9 gene and simultaneous detection using single-strand conformation polymorphism analysis. Leucine 43-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 10850388-1 2000 This study evaluated the catalytic activity of three variants (Ile, Leu, and Thr) at codon 359 of CYP2C9 enzymes expressed in a yeast cDNA expression system, and then established single-strand conformation polymorphism (PCR-SSCP) analysis for simultaneous detection as a screening method. Threonine 77-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10772636-0 2000 Substrate-dependent effect of acetonitrile on human liver microsomal cytochrome P450 2C9 (CYP2C9) activity. acetonitrile 30-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-88 10772636-0 2000 Substrate-dependent effect of acetonitrile on human liver microsomal cytochrome P450 2C9 (CYP2C9) activity. acetonitrile 30-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 10772636-6 2000 The presence of acetonitrile (3%, v/v) gave rise to a lower K(m) and a higher V(max) for diclofenac hydroxylase in both liver microsomes and recombinant CYP2C9 preparations (87 and 52% increase in V(max)/K(m) ratio, respectively). acetonitrile 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). Irbesartan 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). Tolbutamide 56-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). Valsartan 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). candesartan 158-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10877007-5 2000 Losartan and irbesartan inhibited the CYP2C9-associated tolbutamide methylhydroxylation more potently (Ki values 4.1 microM and 24.5 microM), than valsartan, candesartan or eprosartan (Ki values 135 microM, 155 microM and > 1000 microM, respectively). eprosartan 173-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10877007-8 2000 CONCLUSION: All sartans except eprosartan have at least some affinity for CYP2C9, but only losartan has an affinity for CYP2C19. eprosartan 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 10877007-10 2000 This would suggest that the theoretical potential for drug interactions is likely to be quite low, with the possible exceptions of losartan and irbesartan for CYP2C9. Irbesartan 144-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 10725317-13 2000 It is concluded that some of these sulfonamides have to be carefully coadministered with CYP2C9 substrates such as tolbutamide although coadministration of sulfaphenazole needs the greatest care. Sulfonamides 35-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 10806608-1 2000 To assess the effect of tolcapone (an inhibitor of cytochrome P450 [CYP] 2C9 in vitro) on the pharmacokinetics and hypoglycemic effect of the CYP 2C9 substrate tolbutamide, 12 healthy male volunteers were randomized to receive a single dose of tolbutamide 500 mg plus either placebo or tolcapone 200 mg after an overnight fast and 30 minutes after the start of a 6.5-hour 5% glucose infusion (150 mL/h). Tolcapone 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-149 10813596-0 2000 Ontogenesis of CYP2C-dependent arachidonic acid metabolism in the human liver: relationship with sudden infant death syndrome. Arachidonic Acid 31-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-20 10813596-2 2000 To determine the possible consequence of the up-regulation of CYP2C in SIDS, we examined the metabolism of arachidonic acid (AA) an endogenous substrate of CYP2C involved in the physiologic regulation of vascular tone. Arachidonic Acid 107-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-161 10813596-4 2000 In SIDS, the accumulation of CYP2C proteins was associated with a significant increase in the formation of 14,15 and 11,12 diHETE, which were shown to be supported by individually expressed CYP2C8 and 2C9 and HETE1 (presumably 15 HETE). dihete 123-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-34 10813596-5 2000 This increase was markedly inhibited by addition of sulfaphenazole, a selective inhibitor of CYP2C9. Sulfaphenazole 52-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 10813596-6 2000 So, we propose that the higher CYP2C content in SIDS stimulates the production of EETs and diHETEs and might have severe pathologic consequences in children. eets 82-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-36 10813596-6 2000 So, we propose that the higher CYP2C content in SIDS stimulates the production of EETs and diHETEs and might have severe pathologic consequences in children. dihetes 91-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-36 10773015-0 2000 Major role of human liver microsomal cytochrome P450 2C9 (CYP2C9) in the oxidative metabolism of celecoxib, a novel cyclooxygenase-II inhibitor. Celecoxib 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-56 10773015-0 2000 Major role of human liver microsomal cytochrome P450 2C9 (CYP2C9) in the oxidative metabolism of celecoxib, a novel cyclooxygenase-II inhibitor. Celecoxib 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 10773015-3 2000 The treatment of a panel of human liver microsomal samples (n = 16 subjects) with antibodies against CYP2C9 and CYP3A4 inhibited the formation of hydroxy celecoxib by 72 to 92% and 0 to 27%, respectively. Hydroxy Celecoxib 146-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10773015-5 2000 In addition, the formation of hydroxy celecoxib significantly correlated with CYP2C9-selective tolbutamide methyl hydroxylation (r = 0.92, P <. Hydroxy Celecoxib 30-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 10773015-5 2000 In addition, the formation of hydroxy celecoxib significantly correlated with CYP2C9-selective tolbutamide methyl hydroxylation (r = 0.92, P <. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 10773015-10 2000 It is concluded that methyl hydroxylation of celecoxib is primarily catalyzed by human liver microsomal CYP2C9, although CYP3A4 also plays a role. Celecoxib 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 10772937-0 2000 Arginines 97 and 108 in CYP2C9 are important determinants of the catalytic function. Arginine 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 10772937-3 2000 To elucidate the function of the B-C loop we used homology modelling based on the Cyp102 structure in combination with functional studies of mutants using diclofenac as a model substrate for CYP2C9. Diclofenac 155-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 10729206-7 2000 Correlation analysis was performed using individual samples of human liver microsomes, and the best correlation of linoleic acid epoxidation activity was with tolbutamide hydroxylase activity, CYP2C9. Linoleic Acid 115-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 10729206-8 2000 Recombinant CYP2C9 was the most active in linoleic acid epoxygenation, and antibody and chemical inhibition also indicated the importance of CYP2C9. Linoleic Acid 42-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 10725303-6 2000 Tolbutamide hydroxylation tended to be lower in children versus adults (P =.047) (CYP2C9), but did not reach statistical significance after correcting for multiple comparisons. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 10725317-13 2000 It is concluded that some of these sulfonamides have to be carefully coadministered with CYP2C9 substrates such as tolbutamide although coadministration of sulfaphenazole needs the greatest care. Tolbutamide 115-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 10774639-6 2000 Although phenytoin is metabolized predominantly by CYP2C9 with a minor contribution of CYP2C19, patients with the Leu359 variant should be monitored closely when treated with a moderate to high daily dose of phenytoin. Phenytoin 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10853880-0 2000 Lack of differences in diclofenac (a substrate for CYP2C9) pharmacokinetics in healthy volunteers with respect to the single CYP2C9*3 allele. Diclofenac 23-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10853880-1 2000 OBJECTIVES: Evidence exists to suggest that diclofenac is metabolised by CYP2C9. Diclofenac 44-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 10853880-2 2000 The present study was undertaken in order to evaluate the effect of the single CYP2C9*3 variant on drug metabolism using diclofenac as a probe drug. Diclofenac 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 10853880-3 2000 METHODS: A single dose of diclofenac was administered orally to 12 healthy subjects in whom the genotype of CYP2C9 had been determined previously. Diclofenac 26-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 10853880-5 2000 RESULTS: For diclofenac, the following kinetic parameters were observed in the CYP2C9*1/*1 and CYP2C9*1/*3 subjects, respectively (mean +/- SD): apparent oral clearance (ml/kg/h) 355.8 +/- 56.9 and 484.4 +/- 155.3; area under plasma concentration time curve (microg h/ml) 2.7 +/- 0.7 and 1.9 +/- 0.6. Diclofenac 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 10853880-5 2000 RESULTS: For diclofenac, the following kinetic parameters were observed in the CYP2C9*1/*1 and CYP2C9*1/*3 subjects, respectively (mean +/- SD): apparent oral clearance (ml/kg/h) 355.8 +/- 56.9 and 484.4 +/- 155.3; area under plasma concentration time curve (microg h/ml) 2.7 +/- 0.7 and 1.9 +/- 0.6. Diclofenac 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 10853880-6 2000 The formation clearance of 4"-hydroxydiclofenac (ml/kg/h) was 63.6 +/- 19.1 in the CYP2C9*1/*1 subjects compared with 75.9 +/- 27.6 in the CYP2C9*1/*3 subjects. 4'-hydroxydiclofenac 27-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 10853880-6 2000 The formation clearance of 4"-hydroxydiclofenac (ml/kg/h) was 63.6 +/- 19.1 in the CYP2C9*1/*1 subjects compared with 75.9 +/- 27.6 in the CYP2C9*1/*3 subjects. 4'-hydroxydiclofenac 27-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 139-145 10853880-8 2000 CONCLUSION: Since the disposition kinetics of diclofenac does not change in subjects with the single CYP2C9*3 mutant allele, it is suggested that the effects of CYP2C9 polymorphisms on the drug metabolism tend to be substrate specific. Diclofenac 46-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10759690-7 2000 TCL was also a moderate inhibitor of CYP1A2 (Ki = 49 +/- 19 microM) and a weak inhibitor of CYP2C9 (Ki > 75 microM), but its effect on the activities of CYP2E1 (Ki = 584 +/- 48 microM) and CYP3A (> 1000 microM) was marginal. Ticlopidine 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). Sulfaphenazole 26-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). Quinidine 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 10821163-3 2000 The results indicate that sulphaphenazole, quinidine and alpha-naphthoflavone are selective inhibitors of CYP2C9 (IC50 = 0.5-0.7 microM), CYP2D6 (0.3-0.4 microM) and CYP1A (0.05-5 microM) respectively on the basis of the IC50, which are much lower than those of other P450 isoforms (> 10-fold). alpha-naphthoflavone 57-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 10640513-1 2000 CYP2C9 is involved in the metabolism of the oral anticoagulants warfarin, phenprocoumon, and acenocoumarol. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10803683-0 2000 Torsemide metabolism by CYP2C9 variants and other human CYP2C subfamily enzymes. Torsemide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 10718780-4 2000 RESULTS: Amiodarone weakly inhibited CYP2C9, CYP2D6, and CYP3A4-mediated activities with Ki values of 45.1-271.6 microm. Amiodarone 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 10718780-6 2000 The catalytic activities of CYP1A1 (Ki=1.5 microm, alpha=5.7), CYP1A2 (Ki=18.8 microm, alpha=2.6), CYP2C9 (Ki=2.3 microm, alpha=5.9), and CYP2C19 (Ki=15.7 microm, alpha=4.5) were inhibited by desethylamiodarone with mixed type. desethylamiodarone 192-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 10681382-2 2000 Although most studies have implicated CYP2C9 as the exclusive catalyst of hepatic tolbutamide hydroxylation in humans, there is evidence that other CYP2C enzymes (e.g., CYP2C19) may also participate. Tolbutamide 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10681382-7 2000 Monospecific anti-CYP2C9 was also found to inhibit rates of tolbutamide hydroxylation by 93 +/- 4 and 78 +/- 6% in CYP2C19-deficient and CYP2C19-containing human liver microsomes, respectively. Tolbutamide 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 10681382-8 2000 Taken together, our results indicate that both CYP2C9 and CYP2C19 are involved in tolbutamide hydroxylation by human liver microsomes, and that CYP2C19 underlies at least 14 to 22% of tolbutamide metabolism. Tolbutamide 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 10681382-8 2000 Taken together, our results indicate that both CYP2C9 and CYP2C19 are involved in tolbutamide hydroxylation by human liver microsomes, and that CYP2C19 underlies at least 14 to 22% of tolbutamide metabolism. Tolbutamide 184-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 10709776-11 2000 Fluconazole, miconazole and sulfamethoxazole are potent inhibitors of CYP2C9. Fluconazole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 10709776-11 2000 Fluconazole, miconazole and sulfamethoxazole are potent inhibitors of CYP2C9. Miconazole 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 10709776-11 2000 Fluconazole, miconazole and sulfamethoxazole are potent inhibitors of CYP2C9. Sulfamethoxazole 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 10749518-9 2000 Celecoxib is metabolised primarily by the cytochrome P450 (CYP) 2C9 isoenzyme and has an elimination half-life of about 11 hours in healthy individuals. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-67 10749518-14 2000 As celecoxib is metabolised by CYP2C9, increased clinical vigilance is required during the coadministration of other substrates or inhibitors of this enzyme. Celecoxib 3-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 10801223-7 2000 CONCLUSIONS: Although I(inlet,u,max) overestimated the unbound concentration in the liver, the tolbutamide/sulfaphenazole interaction could be successfully predicted by using a fixed value of I(inlet,u,max) indicating that the unbound concentration of sulfaphenazole in the liver after its clinical dose is by far larger than the concentration to inhibit CYP2C9-mediated metabolism and that care should be taken when it is co-administered with drugs that are substrates of CYP2C9. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 355-361 10801223-7 2000 CONCLUSIONS: Although I(inlet,u,max) overestimated the unbound concentration in the liver, the tolbutamide/sulfaphenazole interaction could be successfully predicted by using a fixed value of I(inlet,u,max) indicating that the unbound concentration of sulfaphenazole in the liver after its clinical dose is by far larger than the concentration to inhibit CYP2C9-mediated metabolism and that care should be taken when it is co-administered with drugs that are substrates of CYP2C9. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 473-479 10801223-7 2000 CONCLUSIONS: Although I(inlet,u,max) overestimated the unbound concentration in the liver, the tolbutamide/sulfaphenazole interaction could be successfully predicted by using a fixed value of I(inlet,u,max) indicating that the unbound concentration of sulfaphenazole in the liver after its clinical dose is by far larger than the concentration to inhibit CYP2C9-mediated metabolism and that care should be taken when it is co-administered with drugs that are substrates of CYP2C9. Sulfaphenazole 107-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 355-361 10801223-7 2000 CONCLUSIONS: Although I(inlet,u,max) overestimated the unbound concentration in the liver, the tolbutamide/sulfaphenazole interaction could be successfully predicted by using a fixed value of I(inlet,u,max) indicating that the unbound concentration of sulfaphenazole in the liver after its clinical dose is by far larger than the concentration to inhibit CYP2C9-mediated metabolism and that care should be taken when it is co-administered with drugs that are substrates of CYP2C9. Sulfaphenazole 107-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 473-479 10640508-2 2000 All six compounds reversibly inhibited CYP2D6 (bufuralol 1"-hydroxylation) and CYP2C9 (tolbutamide methyl hydroxylation) activities. Tolbutamide 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 10640508-3 2000 The IC(50) values for the inhibition of CYP2D6 and CYP2C9 for nicardipine were 3 to 9 microM, whereas those for all others ranged from 14 to >150 microM. Nicardipine 62-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10640513-1 2000 CYP2C9 is involved in the metabolism of the oral anticoagulants warfarin, phenprocoumon, and acenocoumarol. Phenprocoumon 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10640513-1 2000 CYP2C9 is involved in the metabolism of the oral anticoagulants warfarin, phenprocoumon, and acenocoumarol. Acenocoumarol 93-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10640513-11 2000 The degree of pharmacokinetic interactions exhibited by oral anticoagulants and lornoxicam is dependent on the respective contribution of CYP2C9 to their total clearance. lornoxicam 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 11144118-4 2000 Colchicine caused an increase of CYP2E1 protein content, colchiceine and N-deacetylcolchiceine induced isoforms CYP2C9, 2E1 and 3A4 whereas colchicoside induced CYP2C9 and 2E1. Colchicine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10805063-9 2000 Nicardipine, benidipine, manidipine and barnidipine competitively inhibited the CYP2C9 and CYP2D6 activities. Nicardipine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 10805063-9 2000 Nicardipine, benidipine, manidipine and barnidipine competitively inhibited the CYP2C9 and CYP2D6 activities. benidipine 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 10805063-9 2000 Nicardipine, benidipine, manidipine and barnidipine competitively inhibited the CYP2C9 and CYP2D6 activities. manidipine 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 10805063-9 2000 Nicardipine, benidipine, manidipine and barnidipine competitively inhibited the CYP2C9 and CYP2D6 activities. mepirodipine 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 10805063-12 2000 Among the human CYP isoforms investigated, the inhibitory effects of 1,4-dihydropyridine calcium antagonists were potent on human CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2D6 as well as CYP3A4. 1,4-dihydropyridine calcium 69-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 10678291-4 2000 Losartan is distinguished from other ARBs by cytochrome P450 (CYP) 3A4- and CYP2C9-mediated biotransformation to its active metabolite EXP-3174. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 10673364-4 2000 Using an RT-competitive PCR (RT-cPCR) with beta-actin as the standard in this study, the constitutive and rifampicin (RFP)-induced expression of CYP3A4, CYP2C9, CYP2E1, and CYP1A2 mRNA in the HepG2 cells could be quantitatively and reproducibly determined. Rifampin 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 10805063-14 2000 CONCLUSIONS: In consideration of the Ki values obtained in the in vitro inhibition study and the concentration of 1,4-dihydropyridine calcium antagonists in human liver, the possibility of in vivo drug interactions of nicardipine and other drugs which are mainly metabolised by CYP2C9 and/or CYP3A4 was suggested. Nicardipine 218-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 278-284 11144118-4 2000 Colchicine caused an increase of CYP2E1 protein content, colchiceine and N-deacetylcolchiceine induced isoforms CYP2C9, 2E1 and 3A4 whereas colchicoside induced CYP2C9 and 2E1. Colchicine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 11144118-4 2000 Colchicine caused an increase of CYP2E1 protein content, colchiceine and N-deacetylcolchiceine induced isoforms CYP2C9, 2E1 and 3A4 whereas colchicoside induced CYP2C9 and 2E1. trimethylcolchicinic acid methyl ether 73-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 11144118-4 2000 Colchicine caused an increase of CYP2E1 protein content, colchiceine and N-deacetylcolchiceine induced isoforms CYP2C9, 2E1 and 3A4 whereas colchicoside induced CYP2C9 and 2E1. colchicoside 140-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10659951-4 2000 Troglitazone (5 microM) significantly inhibited CYP2C8-dependent paclitaxel 6alpha-hydroxylation and CYP2C9-dependent S-warfarin 7-hydroxylation. Troglitazone 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10712746-0 2000 Phenobarbital increases DNA adduct and metabolites formed by ochratoxin A: role of CYP 2C9 and microsomal glutathione-S-transferase. Phenobarbital 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-90 10711629-2 2000 In this study, we examined the extent of thalidomide metabolism by preparations of pooled human microsomes, microsomes containing cloned human CYP isozymes (CYPIA2, CYP2A6, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4), and Hansen"s disease patients. Thalidomide 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 10771452-5 2000 IC50 values for the inhibition of specific probe substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, by ziprasidone, risperidone and 9-hydroxyrisperidone were also determined using human liver microsomes from three subjects. ziprasidone 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 10771452-5 2000 IC50 values for the inhibition of specific probe substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, by ziprasidone, risperidone and 9-hydroxyrisperidone were also determined using human liver microsomes from three subjects. Risperidone 124-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 10771452-5 2000 IC50 values for the inhibition of specific probe substrates for CYP1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A4, by ziprasidone, risperidone and 9-hydroxyrisperidone were also determined using human liver microsomes from three subjects. Paliperidone Palmitate 140-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 10668851-8 2000 CYP2C9 activity was significantly (P < or = .005) increased by 38% +/- 35%, as reflected by the tolbutamide urinary metabolic ratio and oral clearance. Tolbutamide 99-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10611138-0 2000 Stimulation of tolbutamide hydroxylation by acetone and acetonitrile in human liver microsomes and in a cytochrome P-450 2C9-reconstituted system. Tolbutamide 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-124 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. acetonitrile 51-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. acetonitrile 51-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 205-211 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. Acetone 68-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. Acetone 68-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 205-211 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. Tolbutamide 123-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 10611138-3 2000 This study was initiated to evaluate the effect of acetonitrile and acetone, in addition to other organic solvents, on the tolbutamide hydroxylation activity of CYP2C9 in both human liver microsomes and a CYP2C9-reconstituted system. Tolbutamide 123-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 205-211 10611138-4 2000 Both acetonitrile and acetone significantly stimulated the NADPH-dependent tolbutamide hydroxylation by nearly 2- to 3-fold in human liver microsomes and CYP2C9-reconstituted system when incubated at 2 and 4% final solvent concentrations. acetonitrile 5-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 10611138-4 2000 Both acetonitrile and acetone significantly stimulated the NADPH-dependent tolbutamide hydroxylation by nearly 2- to 3-fold in human liver microsomes and CYP2C9-reconstituted system when incubated at 2 and 4% final solvent concentrations. Acetone 22-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 10611138-4 2000 Both acetonitrile and acetone significantly stimulated the NADPH-dependent tolbutamide hydroxylation by nearly 2- to 3-fold in human liver microsomes and CYP2C9-reconstituted system when incubated at 2 and 4% final solvent concentrations. NADP 59-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 10611138-4 2000 Both acetonitrile and acetone significantly stimulated the NADPH-dependent tolbutamide hydroxylation by nearly 2- to 3-fold in human liver microsomes and CYP2C9-reconstituted system when incubated at 2 and 4% final solvent concentrations. Tolbutamide 75-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 10611138-9 2000 Overall, the stimulatory effect of both acetonitrile and acetone on tolbutamide hydroxylation was found to be primarily due to a consistent increase in V(max), whereas K(m) was unchanged in both human liver microsomes and the reconstituted CYP2C9 system. acetonitrile 40-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 10611138-9 2000 Overall, the stimulatory effect of both acetonitrile and acetone on tolbutamide hydroxylation was found to be primarily due to a consistent increase in V(max), whereas K(m) was unchanged in both human liver microsomes and the reconstituted CYP2C9 system. Acetone 57-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 10611138-9 2000 Overall, the stimulatory effect of both acetonitrile and acetone on tolbutamide hydroxylation was found to be primarily due to a consistent increase in V(max), whereas K(m) was unchanged in both human liver microsomes and the reconstituted CYP2C9 system. Tolbutamide 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 10659951-4 2000 Troglitazone (5 microM) significantly inhibited CYP2C8-dependent paclitaxel 6alpha-hydroxylation and CYP2C9-dependent S-warfarin 7-hydroxylation. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 10659951-7 2000 The inhibitory potential of troglitazone (50% inhibition concentration, IC50) was approximately 5 microM for drug oxidations catalyzed by CYP2C9 and CYP2C8 and approximately 20 microM for activities catalyzed by CYP2C19 and CYP3A4 respectively. Troglitazone 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 10562412-0 1999 Structural forms of phenprocoumon and warfarin that are metabolized at the active site of CYP2C9. Phenprocoumon 20-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 10562412-0 1999 Structural forms of phenprocoumon and warfarin that are metabolized at the active site of CYP2C9. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 10562412-4 1999 The conclusion that (S)-warfarin and (S)-phenprocoumon interact with CYP2C9 in very different structural states provides a clear basis for the significant differences observed in their metabolic profiles. Warfarin 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 10562412-4 1999 The conclusion that (S)-warfarin and (S)-phenprocoumon interact with CYP2C9 in very different structural states provides a clear basis for the significant differences observed in their metabolic profiles. Phenprocoumon 37-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 10562412-6 1999 An additional electrostatic binding site also appears to contribute to the orientation of coumarin analogs in the CYP2C9 active site by interacting with the C2-carbonyl group of the coumarin nucleus. coumarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 10562412-6 1999 An additional electrostatic binding site also appears to contribute to the orientation of coumarin analogs in the CYP2C9 active site by interacting with the C2-carbonyl group of the coumarin nucleus. coumarin 182-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 10630892-2 1999 Metabolism of steroid hormones with anabolic properties was studied in vitro using human recombinant CYP3A4, CYP2C9 and 2B6 enzymes. Steroids 14-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-123 10613612-3 1999 In vitro inhibition constants (K(i)) of benzbromarone for (S)-warfarin 7-hydroxylation were determined with use of human CYP2C9 and liver microsomes. Benzbromarone 40-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 10613612-4 1999 The magnitude of changes in the formation clearance for 7-hydroxylation (CLf), the unbound oral clearance (CL(oral,u)), and the oral clearance (CL(oral)) for (S)-warfarin were predicted by equations incorporating the in vitro Ki, the theoretical maximum unbound hepatic benzbromarone concentration, and the fractions of warfarin eliminated through metabolism and of CYP2C9-mediated metabolic reaction susceptible to inhibition by benzbromarone. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 366-372 10613612-7 1999 Benzbromarone was found to be a potent competitive inhibitor (Ki < 0.01 micromol/L) for (S)-warfarin 7-hydroxylation mediated by CYP2C9. Benzbromarone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 10613612-7 1999 Benzbromarone was found to be a potent competitive inhibitor (Ki < 0.01 micromol/L) for (S)-warfarin 7-hydroxylation mediated by CYP2C9. Warfarin 91-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 10613612-9 1999 CONCLUSION: Benzbromarone would intensify anticoagulant response of warfarin through an enantioselective inhibition of CYP2C9-mediated metabolism of pharmacologically more potent (S)-warfarin. Benzbromarone 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 10613612-9 1999 CONCLUSION: Benzbromarone would intensify anticoagulant response of warfarin through an enantioselective inhibition of CYP2C9-mediated metabolism of pharmacologically more potent (S)-warfarin. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 10613612-9 1999 CONCLUSION: Benzbromarone would intensify anticoagulant response of warfarin through an enantioselective inhibition of CYP2C9-mediated metabolism of pharmacologically more potent (S)-warfarin. Warfarin 179-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 10665838-9 1999 In contrast, fluvastatin (mainly metabolized by cytochrome P450-2C9) and pravastatin (eliminated by other metabolic routes) are less subject to this interaction. Fluvastatin 13-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-67 10647906-4 1999 Methylenedioxyphenyl compounds which possess a bulky structure such as 1,4-benzothiazine showed substantial inhibition of S-warfarin 7-hydroxylation catalysed by CYP2C9, S-mephenytoin 4"-hydroxylation by CYP2C19, bufuralol 1"-hydroxylation by CYP2D6, and testosterone 6beta-hydroxylation by CYP3A4. 1,4-benzothiazine 71-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 10647906-4 1999 Methylenedioxyphenyl compounds which possess a bulky structure such as 1,4-benzothiazine showed substantial inhibition of S-warfarin 7-hydroxylation catalysed by CYP2C9, S-mephenytoin 4"-hydroxylation by CYP2C19, bufuralol 1"-hydroxylation by CYP2D6, and testosterone 6beta-hydroxylation by CYP3A4. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 10647906-4 1999 Methylenedioxyphenyl compounds which possess a bulky structure such as 1,4-benzothiazine showed substantial inhibition of S-warfarin 7-hydroxylation catalysed by CYP2C9, S-mephenytoin 4"-hydroxylation by CYP2C19, bufuralol 1"-hydroxylation by CYP2D6, and testosterone 6beta-hydroxylation by CYP3A4. bufuralol 213-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 10647906-4 1999 Methylenedioxyphenyl compounds which possess a bulky structure such as 1,4-benzothiazine showed substantial inhibition of S-warfarin 7-hydroxylation catalysed by CYP2C9, S-mephenytoin 4"-hydroxylation by CYP2C19, bufuralol 1"-hydroxylation by CYP2D6, and testosterone 6beta-hydroxylation by CYP3A4. Testosterone 255-267 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 10627170-0 1999 CYP2C9 is a principal low-affinity phenacetin O-deethylase: fluvoxamine is not a specific CYP1A2 inhibitor. Fluvoxamine 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10586395-1 1999 The objective of this randomized, double-blind, two-period crossover study was to investigate whether concomitant steady-state lansoprazole influences the pharmacokinetics of CYP2C9 substrates using single intravenously dosed phenytoin as a model substrate. Lansoprazole 127-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 10630892-5 1999 When the same formats of CYP2C9 were incubated with the anabolic steroids, no 6beta-hydroxyl metabolites were formed. Steroids 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 10449188-6 1999 Based on microsome studies and the incubations with human hepatocytes and engineered cells, we estimated that in vivo CYP2C9 would be exclusively responsible for the 4" hydroxylation of diclofenac (>99.5%) as well as 5-hydroxydiclofenac (>97%). Diclofenac 186-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Cetirizine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Terfenadine 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Loratadine 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. Astemizole 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-1 1999 Cetirizine, terfenadine, loratadine, astemizole and mizolastine were compared for their ability to inhibit marker activities for CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4 and for some glucuronidation isoenzymes in human liver microsomes. mizolastine 52-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 10597902-4 1999 Furthermore, loratadine activated the CYP2C9-catalyzed tolbutamide hydroxylation (ca. Loratadine 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10597902-4 1999 Furthermore, loratadine activated the CYP2C9-catalyzed tolbutamide hydroxylation (ca. Tolbutamide 55-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 10597902-6 1999 Mizolastine appeared to be a relatively weak and unspecific inhibitor of CYP2E1, CYP2C9, CYP2D6 and CYP3A4 (IC50Ss in the 100 micromolar range). mizolastine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10562462-0 1999 Rapid detection of CYP2C9*3 alleles by real-time fluorescence PCR based on SYBR Green. sybr green 75-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 10562462-1 1999 CYP2C9 catalyzes the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and nonsteroidal anti-inflammatory drugs. Phenytoin 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10562462-1 1999 CYP2C9 catalyzes the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and nonsteroidal anti-inflammatory drugs. Warfarin 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10562462-1 1999 CYP2C9 catalyzes the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and nonsteroidal anti-inflammatory drugs. Tolbutamide 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10562462-1 1999 CYP2C9 catalyzes the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, and nonsteroidal anti-inflammatory drugs. Losartan 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10584064-6 1999 The CYP2C9 model was then used to characterize explicit enzyme complexes with three structurally and chemically diverse substrates: (S)-naproxen, phenytoin, and progesterone. Naproxen 132-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 10584064-6 1999 The CYP2C9 model was then used to characterize explicit enzyme complexes with three structurally and chemically diverse substrates: (S)-naproxen, phenytoin, and progesterone. Phenytoin 146-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 10584064-6 1999 The CYP2C9 model was then used to characterize explicit enzyme complexes with three structurally and chemically diverse substrates: (S)-naproxen, phenytoin, and progesterone. Progesterone 161-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 10870095-6 1999 In vitro inhibition of CYP2C9 by zafirlukast is consistent with clinical studies showing impaired clearance of S-warfarin and enhanced anti-thrombotic effects, although the in vitro IC(50) value is higher than the usual range of clinically relevant plasma concentrations. zafirlukast 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 10870095-6 1999 In vitro inhibition of CYP2C9 by zafirlukast is consistent with clinical studies showing impaired clearance of S-warfarin and enhanced anti-thrombotic effects, although the in vitro IC(50) value is higher than the usual range of clinically relevant plasma concentrations. Sulfur 111-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 10870095-6 1999 In vitro inhibition of CYP2C9 by zafirlukast is consistent with clinical studies showing impaired clearance of S-warfarin and enhanced anti-thrombotic effects, although the in vitro IC(50) value is higher than the usual range of clinically relevant plasma concentrations. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 10870095-7 1999 Zafirlukast deserves further clinical study as an inhibitor of other CYP2C9 substrates such as nonsteroidal anti-inflammatory agents, tolbutamide, phenytoin and mestranol. zafirlukast 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 10449188-6 1999 Based on microsome studies and the incubations with human hepatocytes and engineered cells, we estimated that in vivo CYP2C9 would be exclusively responsible for the 4" hydroxylation of diclofenac (>99.5%) as well as 5-hydroxydiclofenac (>97%). 5-hydroxydiclofenac 220-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 10510155-6 1999 Omeprazole (a CYP2C19 substrate) at high concentrations and sulphaphenazole (a selective inhibitor of CYP2C9) substantially inhibited N-demethylsertraline formation. Sulfaphenazole 60-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 10510154-0 1999 Frequency of cytochrome P450 CYP2C9 variants in a Turkish population and functional relevance for phenytoin. Phenytoin 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 10449188-7 1999 CYP2C9 was exclusively responsible for the formation of 3"-hydroxydiclofenac. 3'-Hydroxydiclofenac 56-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10510154-7 1999 Mean phenytoin serum concentrations at 12 h after dosage were 4.16 mg l-1 (95% CI 3.86-4.46) in carriers of the genotype CYP2C9*1/1, 5.52 mg l-1 (4.66-6.39) in CYP2C9*1/2, and 5.65 mg l-1 (4.86-6.43) in CYP2C9*1/3. Phenytoin 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 10510154-7 1999 Mean phenytoin serum concentrations at 12 h after dosage were 4.16 mg l-1 (95% CI 3.86-4.46) in carriers of the genotype CYP2C9*1/1, 5.52 mg l-1 (4.66-6.39) in CYP2C9*1/2, and 5.65 mg l-1 (4.86-6.43) in CYP2C9*1/3. Phenytoin 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 10510154-7 1999 Mean phenytoin serum concentrations at 12 h after dosage were 4.16 mg l-1 (95% CI 3.86-4.46) in carriers of the genotype CYP2C9*1/1, 5.52 mg l-1 (4.66-6.39) in CYP2C9*1/2, and 5.65 mg l-1 (4.86-6.43) in CYP2C9*1/3. Phenytoin 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 10510154-13 1999 The 12 h serum concentrations after a single phenytoin dose may be used for routine phenotyping of CYP2C9 mediated metabolic clearance and the p-HPPH/P ratios may be even more sensitive indicators of CYP2C9 activity. Phenytoin 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 10510155-6 1999 Omeprazole (a CYP2C19 substrate) at high concentrations and sulphaphenazole (a selective inhibitor of CYP2C9) substantially inhibited N-demethylsertraline formation. desmethylsertraline 134-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 10510155-10 1999 CONCLUSIONS: The polymorphic enzyme CYP2C19 catalyses the high-affinity N-demethylation of sertraline, while CYP2C9 is one of the low-affinity components of this metabolic pathway. Nitrogen 2-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 10510155-10 1999 CONCLUSIONS: The polymorphic enzyme CYP2C19 catalyses the high-affinity N-demethylation of sertraline, while CYP2C9 is one of the low-affinity components of this metabolic pathway. Sertraline 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 10460808-5 1999 Ketoconazole (CYP3A inhibitor) inhibited approximately 40% of the generation of both metabolites, sulfaphenazole (CYP2C inhibitor) inhibited the formation of ND-Z, whereas alpha-naphtoflavone (CYP1A), quinidine (CYP2D6), and chlorzoxazone (CYP2E1) did not affect zopiclone metabolism. Sulfaphenazole 98-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-119 10510156-7 1999 There was some evidence for CYP2C9 playing a minor role in the metabolism of rosiglitazone. Rosiglitazone 77-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 10510156-8 1999 Sulphaphenazole caused limited inhibition (<30%) of both pathways in human liver microsomes and microsomes from cells transfected with CYP2C9 cDNA were able to mediate the metabolism of rosiglitazone, in particular the N-demethylation pathway, albeit at a much slower rate than CYP2C8. Sulfaphenazole 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 10510156-8 1999 Sulphaphenazole caused limited inhibition (<30%) of both pathways in human liver microsomes and microsomes from cells transfected with CYP2C9 cDNA were able to mediate the metabolism of rosiglitazone, in particular the N-demethylation pathway, albeit at a much slower rate than CYP2C8. Rosiglitazone 189-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 10510156-8 1999 Sulphaphenazole caused limited inhibition (<30%) of both pathways in human liver microsomes and microsomes from cells transfected with CYP2C9 cDNA were able to mediate the metabolism of rosiglitazone, in particular the N-demethylation pathway, albeit at a much slower rate than CYP2C8. Nitrogen 147-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 10510156-9 1999 Rosiglitazone caused moderate inhibition of paclitaxel 6alpha-hydroxylase activity (CYP2C8; IC50=18 microm ), weak inhibition of tolbutamide hydroxylase activity (CYP2C9; IC50=50 microm ), but caused no marked inhibition of the other cytochrome P450 activities investigated (CYP1A2, 2A6, 2C9, 2C19, 2D6, 2E1, 3A and 4A). Rosiglitazone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 10460803-6 1999 N-demethylation is primarily mediated by CYP2C9, CYP2C8, and CYP1A2; dihydrodiol formation by CYP2C9 and CYP1A2; deamination by CYP3A4; and side chain oxidation equally by CYP1A2, CYP2C8, CYP2C9, and CYP2C19. Nitrogen 0-1 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 10460803-6 1999 N-demethylation is primarily mediated by CYP2C9, CYP2C8, and CYP1A2; dihydrodiol formation by CYP2C9 and CYP1A2; deamination by CYP3A4; and side chain oxidation equally by CYP1A2, CYP2C8, CYP2C9, and CYP2C19. Nitrogen 0-1 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 10510154-13 1999 The 12 h serum concentrations after a single phenytoin dose may be used for routine phenotyping of CYP2C9 mediated metabolic clearance and the p-HPPH/P ratios may be even more sensitive indicators of CYP2C9 activity. p-hpph 143-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 200-206 10460803-6 1999 N-demethylation is primarily mediated by CYP2C9, CYP2C8, and CYP1A2; dihydrodiol formation by CYP2C9 and CYP1A2; deamination by CYP3A4; and side chain oxidation equally by CYP1A2, CYP2C8, CYP2C9, and CYP2C19. Nitrogen 0-1 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 10460808-7 1999 Recombinant CYP2C8 had the highest enzymatic activity toward zopiclone metabolism into both its metabolites, followed by CYP2C9 and 3A4. zopiclone 61-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 10460803-6 1999 N-demethylation is primarily mediated by CYP2C9, CYP2C8, and CYP1A2; dihydrodiol formation by CYP2C9 and CYP1A2; deamination by CYP3A4; and side chain oxidation equally by CYP1A2, CYP2C8, CYP2C9, and CYP2C19. trans-1,2-dihydro-1,2-naphthalenediol 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 10460803-6 1999 N-demethylation is primarily mediated by CYP2C9, CYP2C8, and CYP1A2; dihydrodiol formation by CYP2C9 and CYP1A2; deamination by CYP3A4; and side chain oxidation equally by CYP1A2, CYP2C8, CYP2C9, and CYP2C19. trans-1,2-dihydro-1,2-naphthalenediol 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 10417492-9 1999 4"-Hydroxymephenytoin formation was inhibited significantly by omeprazole, diethyldithiocarbamate, proguanil, furafylline, diazepam, troleandomycin, and sulphaphenazole (CYP2C9). 4-hydroxymephenytoin 0-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 10503815-5 1999 CYP2C9 is saturated by therapeutic doses of phenytoin, and at steady state both enzymes are probably operant in most people. Phenytoin 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10548450-10 1999 Among the CYP inhibitors examined (alpha-naphthoflavone, sulphaphenazole, omeprazole, quinidine and troleandomycin), sulphaphenazole (a CYP2C9 inhibitor) showed the most potent inhibitory effect on the 5-hydroxylation of S-MTPPA by human liver microsomes. Troleandomycin 100-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 10548450-10 1999 Among the CYP inhibitors examined (alpha-naphthoflavone, sulphaphenazole, omeprazole, quinidine and troleandomycin), sulphaphenazole (a CYP2C9 inhibitor) showed the most potent inhibitory effect on the 5-hydroxylation of S-MTPPA by human liver microsomes. Sulfaphenazole 117-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 10548450-12 1999 When incubated with microsomes containing cDNA-expressed CYP isozymes, S-MTPPA was substantially oxidized to MA6 only by CYP2C9. 2-(4-(3-methyl-2-thienyl)phenyl)propionic acid 71-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 10473927-4 1999 Although the mechanism of the interaction is unclear, it has been postulated that 5-FU interferes with the synthesis of hepatic cytochrome P-450 2C9. Fluorouracil 82-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-148 10417492-9 1999 4"-Hydroxymephenytoin formation was inhibited significantly by omeprazole, diethyldithiocarbamate, proguanil, furafylline, diazepam, troleandomycin, and sulphaphenazole (CYP2C9). Sulfaphenazole 153-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 10383922-6 1999 Azelastine, desmethylazelastine, and 6-hydroxyazelastine competitively inhibited CYP2C9 activity (Ki = 13. azelastine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10383917-5 1999 At the substrate concentration of 20 microM, which approximated the apparent KM value, sulfaphenazole (CYP2C9 inhibitor) and triazolam (CYP3A substrate) reduced the N-demethylation activities by 20 to 35% in human liver microsomes, whereas the inhibition induced by mephenytoin (CYP2C19 substrate) or quinidine (CYP2D6 inhibitor) was marginal. Sulfaphenazole 87-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 10463513-7 1999 Celecoxib is metabolized in the liver by the cytochrome P-450 isozyme CYP2C9, and thus serious drug interactions are possible. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 10383922-6 1999 Azelastine, desmethylazelastine, and 6-hydroxyazelastine competitively inhibited CYP2C9 activity (Ki = 13. desmethylazelastine 12-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10383929-8 1999 Furthermore, allicin formation by human liver microsomes was correlated with p-nitrophenol hydroxylase activity, a marker of CYP2E1, and tolbutamide hydroxylase activity, a marker of CYP2C9. allicin 13-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 10383917-5 1999 At the substrate concentration of 20 microM, which approximated the apparent KM value, sulfaphenazole (CYP2C9 inhibitor) and triazolam (CYP3A substrate) reduced the N-demethylation activities by 20 to 35% in human liver microsomes, whereas the inhibition induced by mephenytoin (CYP2C19 substrate) or quinidine (CYP2D6 inhibitor) was marginal. Triazolam 125-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 10383917-5 1999 At the substrate concentration of 20 microM, which approximated the apparent KM value, sulfaphenazole (CYP2C9 inhibitor) and triazolam (CYP3A substrate) reduced the N-demethylation activities by 20 to 35% in human liver microsomes, whereas the inhibition induced by mephenytoin (CYP2C19 substrate) or quinidine (CYP2D6 inhibitor) was marginal. Mephenytoin 266-277 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 10383917-5 1999 At the substrate concentration of 20 microM, which approximated the apparent KM value, sulfaphenazole (CYP2C9 inhibitor) and triazolam (CYP3A substrate) reduced the N-demethylation activities by 20 to 35% in human liver microsomes, whereas the inhibition induced by mephenytoin (CYP2C19 substrate) or quinidine (CYP2D6 inhibitor) was marginal. Quinidine 301-310 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 10383922-6 1999 Azelastine, desmethylazelastine, and 6-hydroxyazelastine competitively inhibited CYP2C9 activity (Ki = 13. 6-hydroxyazelastine 37-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 10383917-8 1999 In particular, CYP2C19, CYP2B6, CYP2C9-Arg, CYP2D6-Val, and CYP3A4 all showed relatively high activity. Arginine 39-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 10383922-14 1999 The inhibition of CYP2C9, CYP2C19, and CYP3A4 activity by azelastine and its two metabolites might be clinically insignificant. azelastine 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 10383917-10 1999 The results suggest that at least five isoforms of CYP (CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4) are involved in the sertraline N-demethylation in human liver microsomes and that the contribution of any individual isoform does not exceed 40% of overall metabolism. sertraline n 117-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 10413320-1 1999 CYP2C9 is mainly responsible for the metabolic clearance of phenytoin and (S)-warfarin. Phenytoin 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10413320-1 1999 CYP2C9 is mainly responsible for the metabolic clearance of phenytoin and (S)-warfarin. Warfarin 74-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10413320-2 1999 We have shown previously that mutations in the CYP2C9 gene are associated with diminished metabolism of (S)-warfarin, and so we have now studied the metabolism of phenytoin to its primary inactive metabolite, (S)-pHPPH, by these mutant enzymes. Warfarin 104-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 10413320-2 1999 We have shown previously that mutations in the CYP2C9 gene are associated with diminished metabolism of (S)-warfarin, and so we have now studied the metabolism of phenytoin to its primary inactive metabolite, (S)-pHPPH, by these mutant enzymes. Phenytoin 163-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 10413320-2 1999 We have shown previously that mutations in the CYP2C9 gene are associated with diminished metabolism of (S)-warfarin, and so we have now studied the metabolism of phenytoin to its primary inactive metabolite, (S)-pHPPH, by these mutant enzymes. (s)-phpph 209-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 10413320-4 1999 The data demonstrate that the CYP2C9*3 gene product retains only 4-6% of the metabolic efficiency of the wild-type protein, CYP2C9*1, towards phenytoin and (S)-warfarin. Phenytoin 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10413320-4 1999 The data demonstrate that the CYP2C9*3 gene product retains only 4-6% of the metabolic efficiency of the wild-type protein, CYP2C9*1, towards phenytoin and (S)-warfarin. Warfarin 156-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10381752-2 1999 The predominant P-450 enzyme involved in meloxicam metabolism is CYP 2C9, with a minor contribution of CYP 3A4. Meloxicam 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-72 10381752-6 1999 Using expressed cytochrome P-450 enzymes and selective chemical inhibitors of CYP 2C9 and CYP 3A4, it was found that quinidine markedly increased the rate of CYP 3A4-mediated meloxicam hydroxylation but was virtually without effect on CYP 2C9. Quinidine 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 10381752-6 1999 Using expressed cytochrome P-450 enzymes and selective chemical inhibitors of CYP 2C9 and CYP 3A4, it was found that quinidine markedly increased the rate of CYP 3A4-mediated meloxicam hydroxylation but was virtually without effect on CYP 2C9. Quinidine 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 235-242 10381752-6 1999 Using expressed cytochrome P-450 enzymes and selective chemical inhibitors of CYP 2C9 and CYP 3A4, it was found that quinidine markedly increased the rate of CYP 3A4-mediated meloxicam hydroxylation but was virtually without effect on CYP 2C9. Meloxicam 175-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-85 10422890-4 1999 In addition, clozapine may inhibit the activity of CYP2C9 and CYP2C19, and induce CYP1A, CYP2B and CYP3A. Clozapine 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10357778-4 1999 DNA from normal tissue was also evaluated for polymorphisms in cytochrome P450 2C9 (CYP2C9) at two sites, codons 144 (Arg/Cys) and 359 (Ile/Leu), for glutathione S-transferase P1 (GSTP1) at codon 105 and for NAD(P)H:quinone oxidoreductase (NQO1) at codon 187 (Pro/Ser). Arginine 118-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-82 10357778-4 1999 DNA from normal tissue was also evaluated for polymorphisms in cytochrome P450 2C9 (CYP2C9) at two sites, codons 144 (Arg/Cys) and 359 (Ile/Leu), for glutathione S-transferase P1 (GSTP1) at codon 105 and for NAD(P)H:quinone oxidoreductase (NQO1) at codon 187 (Pro/Ser). Arginine 118-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 10357778-4 1999 DNA from normal tissue was also evaluated for polymorphisms in cytochrome P450 2C9 (CYP2C9) at two sites, codons 144 (Arg/Cys) and 359 (Ile/Leu), for glutathione S-transferase P1 (GSTP1) at codon 105 and for NAD(P)H:quinone oxidoreductase (NQO1) at codon 187 (Pro/Ser). nad(p)h 208-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 10348802-4 1999 This investigation determined single-dose disulfiram effects on human CYP 2C9, 2C19, 2D6, and 3A4 activities in vivo. Disulfiram 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-77 10348802-8 1999 CYP2C9, 2C19, 2D6, and 3A4 activities were determined from the tolbutamide metabolic ratio, 4"-hydroxymephenytoin excretion, and dextromethorphan/dextrorphan ratios in urine and midazolam systemic clearance, respectively. Tolbutamide 63-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10348802-8 1999 CYP2C9, 2C19, 2D6, and 3A4 activities were determined from the tolbutamide metabolic ratio, 4"-hydroxymephenytoin excretion, and dextromethorphan/dextrorphan ratios in urine and midazolam systemic clearance, respectively. 4-hydroxymephenytoin 92-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10348802-8 1999 CYP2C9, 2C19, 2D6, and 3A4 activities were determined from the tolbutamide metabolic ratio, 4"-hydroxymephenytoin excretion, and dextromethorphan/dextrorphan ratios in urine and midazolam systemic clearance, respectively. Dextromethorphan 129-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10348802-8 1999 CYP2C9, 2C19, 2D6, and 3A4 activities were determined from the tolbutamide metabolic ratio, 4"-hydroxymephenytoin excretion, and dextromethorphan/dextrorphan ratios in urine and midazolam systemic clearance, respectively. Dextrorphan 146-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10348802-8 1999 CYP2C9, 2C19, 2D6, and 3A4 activities were determined from the tolbutamide metabolic ratio, 4"-hydroxymephenytoin excretion, and dextromethorphan/dextrorphan ratios in urine and midazolam systemic clearance, respectively. Midazolam 178-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 10230773-0 1999 Human CYP2C-mediated stereoselective phenytoin hydroxylation in Japanese: difference in chiral preference of CYP2C9 and CYP2C19. Phenytoin 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 10230773-7 1999 These results, in addition to inhibition experiments with anti-human CYP2C antibody, indicate that phenytoin hydroxylation is mainly catalyzed by CYP2C9. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 10230773-8 1999 Furthermore, CYP2C19 showed limited contribution to phenytoin 4"-hydroxylation with a different chiral preference from CYP2C9. phenytoin 4" 52-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 10357778-4 1999 DNA from normal tissue was also evaluated for polymorphisms in cytochrome P450 2C9 (CYP2C9) at two sites, codons 144 (Arg/Cys) and 359 (Ile/Leu), for glutathione S-transferase P1 (GSTP1) at codon 105 and for NAD(P)H:quinone oxidoreductase (NQO1) at codon 187 (Pro/Ser). Serine 264-267 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 10418968-11 1999 Furafylline, sulfaphenazole, quinidine, and ketoconazole were found to be specific inhibitors of CYP1A2, CYP2C9, CYP2D6, and CYP3A4, respectively. furafylline 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 10418968-11 1999 Furafylline, sulfaphenazole, quinidine, and ketoconazole were found to be specific inhibitors of CYP1A2, CYP2C9, CYP2D6, and CYP3A4, respectively. Sulfaphenazole 13-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 10418968-11 1999 Furafylline, sulfaphenazole, quinidine, and ketoconazole were found to be specific inhibitors of CYP1A2, CYP2C9, CYP2D6, and CYP3A4, respectively. Quinidine 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 10418968-11 1999 Furafylline, sulfaphenazole, quinidine, and ketoconazole were found to be specific inhibitors of CYP1A2, CYP2C9, CYP2D6, and CYP3A4, respectively. Ketoconazole 44-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 10418968-12 1999 Tranylcypromine and diethyldithiocarbamate were found to be less specific, with inhibitory effects towards several CYP isoforms, including CYP2A6, CYP2C9, CYP2C19, and CYP2E1. Tranylcypromine 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10418968-12 1999 Tranylcypromine and diethyldithiocarbamate were found to be less specific, with inhibitory effects towards several CYP isoforms, including CYP2A6, CYP2C9, CYP2C19, and CYP2E1. Ditiocarb 20-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 147-153 10192828-6 1999 Studies using expressed cytochromes showed that ODV was formed by CYP2C9, -2C19, and -2D6. Desvenlafaxine Succinate 48-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10192828-12 1999 Studies using expressed cytochromes showed that NDV was formed by CYP2C9, -2C19, and -3A4. N-desmethylvenlafaxine 48-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10233205-11 1999 CONCLUSIONS: The N-demethylation of methadone in human liver microsomes is not markedly stereoselective, and is mediated mainly by CYP3A4 with the possible involvement of CYP2C9 and CYP2C19. Methadone 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 10233205-8 1999 Sulphaphenazole (CYP2C9) also significantly inhibited (P<0.05) EDDP formation (range 14-25%). Sulfaphenazole 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 10079071-0 1999 Enzymatic determinants of the substrate specificity of CYP2C9: role of B"-C loop residues in providing the pi-stacking anchor site for warfarin binding. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 10233205-8 1999 Sulphaphenazole (CYP2C9) also significantly inhibited (P<0.05) EDDP formation (range 14-25%). 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine 66-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 10197301-0 1999 The effects of fluvastatin, a CYP2C9 inhibitor, on losartan pharmacokinetics in healthy volunteers. Fluvastatin 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10197301-1 1999 Losartan is an angiotensin II receptor antagonist that is metabolized by CYP2C9 and CYP3A4 to a more potent antihypertensive metabolite, E3174. Losartan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 10197301-1 1999 Losartan is an angiotensin II receptor antagonist that is metabolized by CYP2C9 and CYP3A4 to a more potent antihypertensive metabolite, E3174. losartan carboxylic acid 137-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 10197301-8 1999 Inhibition of losartan metabolism appears to require both CYP2C9 and CYP3A4 inhibition. Losartan 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 10375008-6 1999 In comparison with ketoconazole, itraconazole and fluconazole, D0870 was the most potent inhibitor of CYP2C9 activity. Fluconazole 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 10375008-6 1999 In comparison with ketoconazole, itraconazole and fluconazole, D0870 was the most potent inhibitor of CYP2C9 activity. ICI 195739 63-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 10079071-1 1999 Previous modeling efforts have suggested that coumarin ligand binding to CYP2C9 is dictated by electrostatic and pi-stacking interactions with complementary amino acids of the protein. coumarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 10079071-10 1999 Finally, the F114L mutant effected a greater than 100-fold increase in the Ki for inhibition of CYP2C9 activity by sulfaphenazole. Sulfaphenazole 115-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 10079071-11 1999 These data support a role for B"-C helix loop residues F114 and V113 in the hydrophobic binding of warfarin to CYP2C9, and are consistent with pi-stacking to F114 for certain aromatic ligands. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 10073515-0 1999 Association of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 10064574-3 1999 Several enzymes, but mainly CYP2C9, catalyzed fluvastatin metabolism. Fluvastatin 46-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 10064574-4 1999 Only CYP2C9 inhibitors such as sulfaphenazole inhibited the formation of both 6-hydroxy- and N-deisopropyl-fluvastatin. Sulfaphenazole 31-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 10064574-4 1999 Only CYP2C9 inhibitors such as sulfaphenazole inhibited the formation of both 6-hydroxy- and N-deisopropyl-fluvastatin. 6-hydroxy- and n-deisopropyl-fluvastatin 78-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 10064574-5 1999 5-Hydroxy-fluvastatin formation was reduced by compounds that are inhibitors of CYP2C9, CYP3A, or CYP2C8. 5-hydroxy-fluvastatin 0-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 10064574-6 1999 Fluvastatin in turn inhibited CYP2C9-catalyzed tolbutamide and diclofenac hydroxylation with Ki values of 0.3 and 0.5 microM, respectively. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10064574-6 1999 Fluvastatin in turn inhibited CYP2C9-catalyzed tolbutamide and diclofenac hydroxylation with Ki values of 0.3 and 0.5 microM, respectively. Tolbutamide 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10064574-6 1999 Fluvastatin in turn inhibited CYP2C9-catalyzed tolbutamide and diclofenac hydroxylation with Ki values of 0.3 and 0.5 microM, respectively. Diclofenac 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10064574-8 1999 All three fluvastatin metabolites were also formed by recombinant CYP2C9, whereas CYP1A1, CYP2C8, CYP2D6, and CYP3A4 produced only 5-hydroxy-fluvastatin. Fluvastatin 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10064574-12 1999 This data indicates that several human cytochrome P-450 enzymes metabolize fluvastatin with CYP2C9 contributing 50-80%. Fluvastatin 75-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 9925971-5 1999 For formation of M2, both a rabbit CYP3A polyclonal antibody (110 microliter/mg microsomal protein) and ketoconazole (2 micromol/l), a CYP3A inhibitor, showed about 50% inhibitory effects; other specific inhibitors of CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP2E1 showed no significant effects. Ketoconazole 104-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 234-240 10073515-1 1999 BACKGROUND: The cytochrome P450 CYP2C9 is responsible for the metabolism of S-warfarin. Warfarin 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 10073515-4 1999 We have studied the effect of CYP2C9 polymorphism on the in-vivo warfarin dose requirement. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 10073515-8 1999 FINDINGS: The odds ratio for individuals with a low warfarin dose requirement having one or more CYP2C9 variant alleles compared with the normal population was 6.21 (95% CI 2.48-15.6). Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 10073515-10 1999 INTERPRETATION: We have shown that there is a strong association between CYP2C9 variant alleles and low warfarin dose requirement. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 10073515-11 1999 CYP2C9 genotyping may identify a subgroup of patients who have difficulty at induction of warfarin therapy and are potentially at a higher risk of bleeding complications. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9929518-13 1999 Enzyme kinetics suggest that nifedipine is a noncompetitive-type inhibitor of CYP2C9-mediated catalytic activities. Nifedipine 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 10027801-4 1999 Cytochrome P450 2C9, which catalyzes the major route of oxidative metabolism of diclofenac to produce 4"-hydroxydiclofenac, did not appear to be responsible for the formation of the protein adducts, because sulfaphenazole, an inhibitor of this enzyme, did not affect protein adduct formation. Diclofenac 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 10027801-4 1999 Cytochrome P450 2C9, which catalyzes the major route of oxidative metabolism of diclofenac to produce 4"-hydroxydiclofenac, did not appear to be responsible for the formation of the protein adducts, because sulfaphenazole, an inhibitor of this enzyme, did not affect protein adduct formation. 4'-hydroxydiclofenac 102-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 10190649-2 1999 Despite the widespread use of tolbutamide in Asian populations, the pharmacokinetics of tolbutamide, a CYP2C9 substrate, have not been described in ethnic Chinese. Tolbutamide 88-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 9929510-3 1999 Methanol did not substantially inhibit (</=10%) any of the activities at 0.3%, but did inhibit CYP1A1, CYP2B6, CYP2C9, and CYP2D6 by 12 to 26% at 1%. Methanol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 9929518-14 1999 Finally, only microsomes containing recombinant human liver CYP2C9 were capable of oxidizing irbesartan. Irbesartan 93-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 9929510-10 1999 Acetonitrile also increased CYP2C9 activity by 10 to 15% above control values at 1 to 3% solvent. acetonitrile 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 9929518-7 1999 769) with tolbutamide (CYP2C9 substrate) 4-methyl-hydroxylation. Tolbutamide 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 9929518-15 1999 These results provide evidence that CYP2C9 plays a major role in irbesartan oxidation. Irbesartan 65-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 9929518-8 1999 Oxidation of irbesartan was markedly inhibited by sulfaphenazole (CYP2C9 inhibitor), but not by any of several other CYP inhibitors. Irbesartan 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 9929518-8 1999 Oxidation of irbesartan was markedly inhibited by sulfaphenazole (CYP2C9 inhibitor), but not by any of several other CYP inhibitors. Sulfaphenazole 50-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 10192756-0 1999 Inhibition of CYP2C9 by selective serotonin reuptake inhibitors: in vitro studies with tolbutamide and (S)-warfarin using human liver microsomes. Warfarin 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 9929518-9 1999 In the same manner, both tolbutamide and warfarin (CYP2C9 substrates), were competitive-type inhibitors of irbesartan oxidation with Ki values of 500 and 30 microM, respectively. Tolbutamide 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 9929518-9 1999 In the same manner, both tolbutamide and warfarin (CYP2C9 substrates), were competitive-type inhibitors of irbesartan oxidation with Ki values of 500 and 30 microM, respectively. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 9929518-9 1999 In the same manner, both tolbutamide and warfarin (CYP2C9 substrates), were competitive-type inhibitors of irbesartan oxidation with Ki values of 500 and 30 microM, respectively. Irbesartan 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 10192756-1 1999 OBJECTIVE: To investigate the in vitro potential of selective serotonin reuptake inhibitors (SSRIs) to inhibit two CYP2C9-catalysed reactions, tolbutamide 4-methylhydroxylation and (S)-warfarin 7-hydroxylation. Tolbutamide 143-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 10192756-10 1999 This is consistent with in vivo data indicating that amongst the SSRIs, fluvoxamine has the greatest potential for inhibiting CYP2C9-mediated drug metabolism. Fluvoxamine 72-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 10208645-7 1999 An additional blood sample was obtained from this individual to determine if he possessed any of the known CYP2C9 or CYP2C19 allelic variants that would account for his poor clearance of the CYP2C9 substrates (phenytoin and glipizide) compared with the CYP3A4 (nifedipine) and CYP2D6 (chlorpheniramine) substrates. Nifedipine 261-271 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 10048600-3 1999 In the present study, in vitro metabolism of risperidone (100 microM) was investigated using the recombinant human cytochrome P450 (CYP) enzymes CYP1A1, CYP1A2, CYP2C8, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 supplemented with an NADPH-generating system. Risperidone 45-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 10048600-3 1999 In the present study, in vitro metabolism of risperidone (100 microM) was investigated using the recombinant human cytochrome P450 (CYP) enzymes CYP1A1, CYP1A2, CYP2C8, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4 and CYP3A5 supplemented with an NADPH-generating system. Risperidone 45-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 184-190 10208645-7 1999 An additional blood sample was obtained from this individual to determine if he possessed any of the known CYP2C9 or CYP2C19 allelic variants that would account for his poor clearance of the CYP2C9 substrates (phenytoin and glipizide) compared with the CYP3A4 (nifedipine) and CYP2D6 (chlorpheniramine) substrates. Chlorpheniramine 285-301 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Phenytoin 81-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Phenytoin 81-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Warfarin 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Warfarin 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Tolbutamide 104-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Tolbutamide 104-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Losartan 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Losartan 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Torsemide 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9920790-1 1999 Cytochrome P450 2C9 (CYP2C9) catalysis the metabolism of important drugs such as phenytoin, S-warfarin, tolbutamide, losartan, torasemide, and nonsteroidal anti-inflammatory drugs. Torsemide 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9860067-0 1998 The effects of genetic polymorphisms of CYP2C9 and CYP2C19 on phenytoin metabolism in Japanese adult patients with epilepsy: studies in stereoselective hydroxylation and population pharmacokinetics. Phenytoin 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 10574228-5 1999 Ginsenoside Rd produced weak inhibitory activity against the surrogate substrates for CYP3A4 and CYP2D6 and even weaker inhibitory activity against the surrogate substrates for CYP2C19 and CYP2C9. Ginsenosides 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 189-195 10574228-7 1999 Ginsenoside Rc produced an increase in the activity of CYP2C9 (70% at 200 uM) and ginsenoside Rf produced an increase in the activity of CYP3A4 (54% at 200 uM). ginsenoside Rc 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 10078840-6 1999 Furafylline, sulphaphenazole, omeprazole, quinidine and ketoconazole were identified as specific markers for the respective CYP1A2 (IC50 = 6 microM), CYP2C9 (0.7 microM), CYP2C19 (6 microM), CYP2D6 (0.02 microM) and CYP3A4 (0.2 microM) inhibition screens. Ketoconazole 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 10208642-0 1999 The effect of genetic polymorphisms in CYP2C9 on sulphamethoxazole N-hydroxylation. sulphamethoxazole n 49-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 10208642-1 1999 Sulphamethoxazole undergoes CYP2C9-mediated bioactivation to a hydroxylamine. Sulfamethoxazole 0-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 10208642-1 1999 Sulphamethoxazole undergoes CYP2C9-mediated bioactivation to a hydroxylamine. Hydroxylamine 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 10208642-2 1999 In this study, we investigated the effect of the CYP2C9Arg144 to Cys (CYP2C9*2) and CYP2C9Ile359 to Leu (CYP2C9*3) polymorphisms on sulphamethoxazole N-hydroxylation. Sulfamethoxazole 132-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 10208642-5 1999 Microsomes prepared from the cell line expressing the allelic variants CYP2C9-Cys144 and CYP2C9-Leu359 displayed a threefold and 20-fold decrease in intrinsic clearance (Cl(int)) for sulphamethoxazole, respectively, when compared with the wild-type, CYP2C9-Arg144. Sulfamethoxazole 183-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 10208642-5 1999 Microsomes prepared from the cell line expressing the allelic variants CYP2C9-Cys144 and CYP2C9-Leu359 displayed a threefold and 20-fold decrease in intrinsic clearance (Cl(int)) for sulphamethoxazole, respectively, when compared with the wild-type, CYP2C9-Arg144. Sulfamethoxazole 183-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 10208642-5 1999 Microsomes prepared from the cell line expressing the allelic variants CYP2C9-Cys144 and CYP2C9-Leu359 displayed a threefold and 20-fold decrease in intrinsic clearance (Cl(int)) for sulphamethoxazole, respectively, when compared with the wild-type, CYP2C9-Arg144. Sulfamethoxazole 183-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 10208642-11 1999 The CYP2C9*2 and CYP2C9*3 polymorphisms may have some influence on the bioactivation of sulphamethoxazole, particularly in individuals who are homozygous mutants, and this could act as a protective factor against sulphamethoxazole hypersensitivity. Sulfamethoxazole 88-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 10208642-11 1999 The CYP2C9*2 and CYP2C9*3 polymorphisms may have some influence on the bioactivation of sulphamethoxazole, particularly in individuals who are homozygous mutants, and this could act as a protective factor against sulphamethoxazole hypersensitivity. Sulfamethoxazole 88-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 10208642-11 1999 The CYP2C9*2 and CYP2C9*3 polymorphisms may have some influence on the bioactivation of sulphamethoxazole, particularly in individuals who are homozygous mutants, and this could act as a protective factor against sulphamethoxazole hypersensitivity. Sulfamethoxazole 213-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 10208642-11 1999 The CYP2C9*2 and CYP2C9*3 polymorphisms may have some influence on the bioactivation of sulphamethoxazole, particularly in individuals who are homozygous mutants, and this could act as a protective factor against sulphamethoxazole hypersensitivity. Sulfamethoxazole 213-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 10208645-0 1999 Pharmacokinetics of chlorpheniramine, phenytoin, glipizide and nifedipine in an individual homozygous for the CYP2C9*3 allele. Chlorpheniramine 20-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 10208645-0 1999 Pharmacokinetics of chlorpheniramine, phenytoin, glipizide and nifedipine in an individual homozygous for the CYP2C9*3 allele. Nifedipine 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 10208645-4 1999 A male Caucasian who participated in six bioavailability studies in our laboratory over a period of several years showed extremely low clearance of two drugs: phenytoin and glipizide (both substrates of CYP2C9), but not for nifedipine (a CYP3A4 substrate) and chlorpheniramine (a CYP2D6 substrate). Phenytoin 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 10208645-4 1999 A male Caucasian who participated in six bioavailability studies in our laboratory over a period of several years showed extremely low clearance of two drugs: phenytoin and glipizide (both substrates of CYP2C9), but not for nifedipine (a CYP3A4 substrate) and chlorpheniramine (a CYP2D6 substrate). Glipizide 173-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 10208645-4 1999 A male Caucasian who participated in six bioavailability studies in our laboratory over a period of several years showed extremely low clearance of two drugs: phenytoin and glipizide (both substrates of CYP2C9), but not for nifedipine (a CYP3A4 substrate) and chlorpheniramine (a CYP2D6 substrate). Chlorpheniramine 260-276 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 203-209 10208645-7 1999 An additional blood sample was obtained from this individual to determine if he possessed any of the known CYP2C9 or CYP2C19 allelic variants that would account for his poor clearance of the CYP2C9 substrates (phenytoin and glipizide) compared with the CYP3A4 (nifedipine) and CYP2D6 (chlorpheniramine) substrates. Phenytoin 210-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 10208645-7 1999 An additional blood sample was obtained from this individual to determine if he possessed any of the known CYP2C9 or CYP2C19 allelic variants that would account for his poor clearance of the CYP2C9 substrates (phenytoin and glipizide) compared with the CYP3A4 (nifedipine) and CYP2D6 (chlorpheniramine) substrates. Phenytoin 210-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 10208645-7 1999 An additional blood sample was obtained from this individual to determine if he possessed any of the known CYP2C9 or CYP2C19 allelic variants that would account for his poor clearance of the CYP2C9 substrates (phenytoin and glipizide) compared with the CYP3A4 (nifedipine) and CYP2D6 (chlorpheniramine) substrates. Glipizide 224-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 10078840-6 1999 Furafylline, sulphaphenazole, omeprazole, quinidine and ketoconazole were identified as specific markers for the respective CYP1A2 (IC50 = 6 microM), CYP2C9 (0.7 microM), CYP2C19 (6 microM), CYP2D6 (0.02 microM) and CYP3A4 (0.2 microM) inhibition screens. furafylline 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 9866708-3 1998 CYP2C9 converted arachidonic and linoleic acids to epoxides/diols and monohydroxy fatty acids. arachidonic 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9866708-3 1998 CYP2C9 converted arachidonic and linoleic acids to epoxides/diols and monohydroxy fatty acids. Linoleic Acids 33-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9866708-3 1998 CYP2C9 converted arachidonic and linoleic acids to epoxides/diols and monohydroxy fatty acids. Epoxy Compounds 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9866708-3 1998 CYP2C9 converted arachidonic and linoleic acids to epoxides/diols and monohydroxy fatty acids. diols 60-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9866708-3 1998 CYP2C9 converted arachidonic and linoleic acids to epoxides/diols and monohydroxy fatty acids. monohydroxy fatty acids 70-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9866708-5 1998 Incubation of CYP2C9 under oxygen-18 gas showed that all HETEs had incorporated oxygen-18 to the same degree. Oxygen 27-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 9866708-5 1998 Incubation of CYP2C9 under oxygen-18 gas showed that all HETEs had incorporated oxygen-18 to the same degree. Oxygen 80-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 9866708-6 1998 Chiral HPLC showed that CYP2C9 formed 15R-HETE (72% of the R enantiomer), 13S-HETE (90%), and 11R-HETE (57%). 15R-HETE 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 9866708-6 1998 Chiral HPLC showed that CYP2C9 formed 15R-HETE (72% of the R enantiomer), 13S-HETE (90%), and 11R-HETE (57%). 13-hydroxyeicosatetraenoic acid 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 9860067-1 1998 PURPOSE: The aim of this study was to clarify the effects of genetic polymorphisms of cytochrome P450 (CYP) 2C9 and 2C19 on the metabolism of phenytoin (PHT). Phenytoin 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-120 9860067-1 1998 PURPOSE: The aim of this study was to clarify the effects of genetic polymorphisms of cytochrome P450 (CYP) 2C9 and 2C19 on the metabolism of phenytoin (PHT). Phenytoin 153-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-120 9866708-6 1998 Chiral HPLC showed that CYP2C9 formed 15R-HETE (72% of the R enantiomer), 13S-HETE (90%), and 11R-HETE (57%). 11-hydroxy-5,8,12,14-eicosatetraenoic acid 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 9864282-15 1998 We conclude that CYP2C9 and possibly CYP2C19 and important isoforms catalysing the initial hydroxylation of desogestrel. Desogestrel 108-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 9916601-9 1998 Sildenafil is cleared predominantly by the hepatic microsomal isoenzymes CYP3A4 (major route) and CYP2C9 (minor route). Sildenafil Citrate 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 9864282-5 1998 Desogestrel was metabolised by microsomes from lymphoblasts containing cDNA-expressed CYP2C9 and CYP2C19 to 3alpha-hydroxydesogestrel with small amounts of 3beta-hydroxydesogestrel also being observed. Desogestrel 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 9864282-9 1998 Sulfaphenazole potently inhibited 3alpha-hydroxydesogestrel formation by CYP2C9 microsomes with a Ki value of 0.91 microM. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 9864282-9 1998 Sulfaphenazole potently inhibited 3alpha-hydroxydesogestrel formation by CYP2C9 microsomes with a Ki value of 0.91 microM. 3alpha-Hydroxydesogestrel 34-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 9864282-11 1998 Sulfaphenazole partially inhibited 3alpha-hydroxydesogestrel and 3-ketodesogestrel formation in human liver microsomes indicating a possible in vivo role for CYP2C9. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 9840430-3 1998 In vitro metabolism of clozapine was investigated using human CYP1A1, CYP1A2, CYP2C8, CYP2E1, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4, CYP3A5 and FMO3 supplemented with an NADPH generating system. Clozapine 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 9840430-3 1998 In vitro metabolism of clozapine was investigated using human CYP1A1, CYP1A2, CYP2C8, CYP2E1, CYP2C9-arg144, CYP2C9-cys144, CYP2C19, CYP2D6, CYP3A4, CYP3A5 and FMO3 supplemented with an NADPH generating system. Clozapine 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 9763401-7 1998 Ketoconazole (2.0 microM), a CYP3A-selective inhibitor, inhibited the hydroxylation of both compounds by 53-67%, and sulfaphenazole (CYP2C9-selective) decreased activity by 10-20%. Sulfaphenazole 117-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 9735164-0 1998 Identification of residues 286 and 289 as critical for conferring substrate specificity of human CYP2C9 for diclofenac and ibuprofen. Diclofenac 108-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9812178-16 1998 Since ritonavir is also an inducer of several metabolising enzymes [CYP1A4, glucuronosyl transferase (GT), and possibly CYP2C9 and CYP2C19], the magnitude of drug interactions is difficult to predict, particularly for drugs that are metabolised by multiple enzymes or have low intrinsic clearance by CYP3A. Ritonavir 6-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 9825828-0 1998 Comparisons between in-vitro and in-vivo metabolism of (S)-warfarin: catalytic activities of cDNA-expressed CYP2C9, its Leu359 variant and their mixture versus unbound clearance in patients with the corresponding CYP2C9 genotypes. Warfarin 55-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 9825828-0 1998 Comparisons between in-vitro and in-vivo metabolism of (S)-warfarin: catalytic activities of cDNA-expressed CYP2C9, its Leu359 variant and their mixture versus unbound clearance in patients with the corresponding CYP2C9 genotypes. Warfarin 55-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 213-219 9825828-5 1998 Consequently, there was a significant correlation (r = 0.99, P < 0.05) between the in-vitro Cl(int) for (S)-warfarin 7-hydroxylation and the in-vivo Cl(po,u) for (S)-warfarin in relation to the CYP2C9*3 polymorphism. Warfarin 107-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 9825828-6 1998 In conclusion, the in-vitro model for human CYP2C9*3 polymorphism using recombinant cytochrome P450 proteins would serve as a useful means for predicting changes in in-vivo kinetics for (S)-warfarin and possibly other CYP2C9 substrates in relation to CYP2C9*3 polymorphism. Warfarin 186-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9825830-5 1998 Haloperidol and CPHP formation rates in the nine liver preparations were significantly correlated with dextromethorphan N-demethylase activity (a marker of CYP3A4 activity), but not with the CYP2D6, CYP1A2 and CYP2C9 activity. Haloperidol 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 210-216 9825830-5 1998 Haloperidol and CPHP formation rates in the nine liver preparations were significantly correlated with dextromethorphan N-demethylase activity (a marker of CYP3A4 activity), but not with the CYP2D6, CYP1A2 and CYP2C9 activity. 4-(4'-chlorophenyl)-4-piperidinol 16-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 210-216 9735164-0 1998 Identification of residues 286 and 289 as critical for conferring substrate specificity of human CYP2C9 for diclofenac and ibuprofen. Ibuprofen 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9735164-1 1998 Specificity of human CYP2C9 for two substrates, diclofenac and ibuprofen, was studied using chimeras and site-directed mutants of CYP2C9 and the highly related CYP2C19 expressed in Escherichia coli. Diclofenac 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9804052-5 1998 In the treatment of transplant patients, from a drug interaction perspective, pravastatin, which is not significantly metabolized by CYP enzymes, and fluvastatin, presumably a CYP2C9 substrate, compare favorably with the other statins for which the major metabolic pathways are catalyzed by CYP3A. Fluvastatin 150-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 9735164-1 1998 Specificity of human CYP2C9 for two substrates, diclofenac and ibuprofen, was studied using chimeras and site-directed mutants of CYP2C9 and the highly related CYP2C19 expressed in Escherichia coli. Ibuprofen 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9735164-1 1998 Specificity of human CYP2C9 for two substrates, diclofenac and ibuprofen, was studied using chimeras and site-directed mutants of CYP2C9 and the highly related CYP2C19 expressed in Escherichia coli. Ibuprofen 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 9735164-4 1998 The regiospecificity of this construct for metabolism of ibuprofen differed from that of CYP2C9 by favoring 2-hydroxylation over 3-hydroxylation. Ibuprofen 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 9735164-5 1998 A CYP2C9 construct containing residues 228-340 of CYP2C19 lacked both diclofenac and ibuprofen hydroxylase activities. Diclofenac 70-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 2-8 9735164-6 1998 When residues 228-282 (containing SRS 3) of CYP2C9 were replaced by those of CYP2C19, the chimera retained appreciable activity for diclofenac and ibuprofen, and tolbutamide activity was inhibited by a specific CYP2C9 inhibitor, sulfaphenazole. Diclofenac 132-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9735164-6 1998 When residues 228-282 (containing SRS 3) of CYP2C9 were replaced by those of CYP2C19, the chimera retained appreciable activity for diclofenac and ibuprofen, and tolbutamide activity was inhibited by a specific CYP2C9 inhibitor, sulfaphenazole. Ibuprofen 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9735164-6 1998 When residues 228-282 (containing SRS 3) of CYP2C9 were replaced by those of CYP2C19, the chimera retained appreciable activity for diclofenac and ibuprofen, and tolbutamide activity was inhibited by a specific CYP2C9 inhibitor, sulfaphenazole. Tolbutamide 162-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9735164-6 1998 When residues 228-282 (containing SRS 3) of CYP2C9 were replaced by those of CYP2C19, the chimera retained appreciable activity for diclofenac and ibuprofen, and tolbutamide activity was inhibited by a specific CYP2C9 inhibitor, sulfaphenazole. Sulfaphenazole 229-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9735164-13 1998 Therefore residues 286 and 289 of CYP2C9 are important in conferring specificity for diclofenac and ibuprofen. Diclofenac 85-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 9735164-13 1998 Therefore residues 286 and 289 of CYP2C9 are important in conferring specificity for diclofenac and ibuprofen. Ibuprofen 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 9733665-7 1998 Co-incubation of liver microsomes with methoxychlor and various P450 isoform-selective inhibitors suggested involvement of several P450s in mono-O-demethylation, including CYP1A2, CYP2A6, CYP2C9, and CYP2C19. Methoxychlor 39-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 9811160-0 1998 Multiple human cytochromes contribute to biotransformation of dextromethorphan in-vitro: role of CYP2C9, CYP2C19, CYP2D6, and CYP3A. Dextromethorphan 62-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9764927-3 1998 Propofol produced inhibition of CYP1A2 (phenacetin O-deethylation), CYP2C9 (tolbutamide 4"-hydroxylation), CYP2D6 (dextromethorphan O-demethylation) and CYP3A4 (testosterone 6beta-hydroxylation) activities with IC50 = 40, 49, 213 and 32 microM respectively. Propofol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 9811160-4 1998 Dextrorphan was formed from dextromethorphan by microsomes from cDNA-transfected lymphoblastoid cells expressing CYP2C9, -2C19, and -2D6 but not by those expressing CYP1A2, -2E1 or -3A4. Dextrorphan 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 9811160-8 1998 3-Methoxymorphinan was formed by microsomes from cDNA-transfected lymphoblastoid cells expressing CYP2C9, -2C19, -2D6, and -3A4, but not by those expressing CYP1A2 or -2E1. 3-methoxymorphinan 0-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 9811160-11 1998 After adjustment for relative abundance, CYP3A4 was identified as the dominant enzyme mediating 3-methoxymorphinan formation from dextromethorphan, although CYP2C9 and -2C19 were estimated to contribute to 3-methoxymorphinan formation, particularly at low substrate concentrations. 3-methoxymorphinan 206-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 9698292-0 1998 Human N-demethylation of (S)-mephenytoin by cytochrome P450s 2C9 and 2B6. Mephenytoin 25-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-72 9698292-5 1998 Of the recombinant P450 isoforms tested, only CYP2B6 and CYP2C9 formed nirvanol from (S)-MP. ethylphenylhydantoin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Ibuprofen 124-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Nortriptyline 174-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Caffeine 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Acetaminophen 211-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Theophylline 42-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Lidocaine 250-259 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Midazolam 261-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9867310-3 1998 The main probe drugs include caffeine and theophylline, whose metabolism is catalysed by CYP1A2, tolbutamide, phenytoin and ibuprofen, catalysed by CYP2C9, amitriptyline and nortriptyline, catalysed by CYP2C19, acetaminophen, catalysed by CYP2E1 and lidocaine, midazolam and terfenadine, catalysed by 3A3/4. Terfenadine 275-286 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 9698079-0 1998 Comparative studies on the catalytic roles of cytochrome P450 2C9 and its Cys- and Leu-variants in the oxidation of warfarin, flurbiprofen, and diclofenac by human liver microsomes. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 9698079-0 1998 Comparative studies on the catalytic roles of cytochrome P450 2C9 and its Cys- and Leu-variants in the oxidation of warfarin, flurbiprofen, and diclofenac by human liver microsomes. Flurbiprofen 126-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 9698079-0 1998 Comparative studies on the catalytic roles of cytochrome P450 2C9 and its Cys- and Leu-variants in the oxidation of warfarin, flurbiprofen, and diclofenac by human liver microsomes. Diclofenac 144-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 9698079-7 1998 Activities for the oxidation of these CYP2C9 substrates in humans with heterozygous Leu359 allele is likely to be dependent on the levels of expression of each of the wild- and Leu-variants in the livers. Leucine 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 9698079-8 1998 However, one of the humans with a heterozygous Leu allele was found to have very low activities towards the oxidation of CYP2C9 substrates. Leucine 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 9578596-0 1998 Characterization of CYP2C19 and CYP2C9 from human liver: respective roles in microsomal tolbutamide, S-mephenytoin, and omeprazole hydroxylations. Tolbutamide 88-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9755458-7 1998 Recently, an allelic variant of CYP2C9 that causes substitution of Leu359 for Ile359 has been shown to be associated with the decreased metabolic clearance of various therapeutic agents including warfarin, tolbutamide and phenytoin. Warfarin 196-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9755458-7 1998 Recently, an allelic variant of CYP2C9 that causes substitution of Leu359 for Ile359 has been shown to be associated with the decreased metabolic clearance of various therapeutic agents including warfarin, tolbutamide and phenytoin. Tolbutamide 206-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9755458-7 1998 Recently, an allelic variant of CYP2C9 that causes substitution of Leu359 for Ile359 has been shown to be associated with the decreased metabolic clearance of various therapeutic agents including warfarin, tolbutamide and phenytoin. Phenytoin 222-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9663807-3 1998 CYP2C9 activity in vivo is inducible by rifampicin. Rifampin 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9663807-4 1998 Evidence suggests that CYP2C9 substrates may also be induced variably by carbamazepine, ethanol and phenobarbitone. Carbamazepine 73-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 9663807-4 1998 Evidence suggests that CYP2C9 substrates may also be induced variably by carbamazepine, ethanol and phenobarbitone. Ethanol 88-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 9663807-4 1998 Evidence suggests that CYP2C9 substrates may also be induced variably by carbamazepine, ethanol and phenobarbitone. Phenobarbital 100-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 9663807-7 1998 Polymorphisms in the coding region of the CYP2C9 gene produce variants at amino acid residues 144 (Arg144Cys) and 359 (Ile359Leu) of the CYP2C9 protein. ile359leu 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9663807-7 1998 Polymorphisms in the coding region of the CYP2C9 gene produce variants at amino acid residues 144 (Arg144Cys) and 359 (Ile359Leu) of the CYP2C9 protein. ile359leu 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 9616194-5 1998 Trazodone (100 microM) was then incubated with microsomes from cells expressing human CYP1A1, CYP1A2, CYP2C8, CYP2C9arg, CYP2C9cys, CYP2C19, CYP2D6, or CYP3A4. Trazodone 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 9616194-5 1998 Trazodone (100 microM) was then incubated with microsomes from cells expressing human CYP1A1, CYP1A2, CYP2C8, CYP2C9arg, CYP2C9cys, CYP2C19, CYP2D6, or CYP3A4. Trazodone 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 9578596-3 1998 To that end, we purified CYP2C19, CYP2C9, and CYP2C8 from human liver samples using conventional chromatographic techniques and examined their capacity to oxidize S-mephenytoin, omeprazole, and tolbutamide. Mephenytoin 163-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 9578596-3 1998 To that end, we purified CYP2C19, CYP2C9, and CYP2C8 from human liver samples using conventional chromatographic techniques and examined their capacity to oxidize S-mephenytoin, omeprazole, and tolbutamide. Tolbutamide 194-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 9578596-6 1998 The kinetic parameters of CYP2C19-mediated tolbutamide hydroxylation (Km = 650 microM, Vmax = 3.71 min-1) somewhat resembled those of the CYP2C9-catalyzed reaction (Km = 178-407 microM, Vmax = 2.95-7.08 min-1). Tolbutamide 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 9578596-0 1998 Characterization of CYP2C19 and CYP2C9 from human liver: respective roles in microsomal tolbutamide, S-mephenytoin, and omeprazole hydroxylations. Mephenytoin 101-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9578596-11 1998 A significant relationship was likewise observed between microsomal tolbutamide hydroxylation and CYP2C9 content (r = 0.664; P < 0.02) but not with CYP2C19 content (r = 0.393; P = 0.184). Tolbutamide 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 9578596-0 1998 Characterization of CYP2C19 and CYP2C9 from human liver: respective roles in microsomal tolbutamide, S-mephenytoin, and omeprazole hydroxylations. Omeprazole 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9630825-0 1998 Metabolism of warfarin enantiomers in Japanese patients with heart disease having different CYP2C9 and CYP2C19 genotypes. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 9630825-1 1998 OBJECTIVE: To determine whether genetic polymorphism of cytochrome P450 (CYP) 2C9 affects the in vivo metabolism of warfarin enantiomers. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-81 9630825-11 1998 CONCLUSION: Heterozygotes for CYP2C9 I1e359/Leu allele have reduced in vivo metabolism of (S)-warfarin but not (R)-warfarin. Leucine 44-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 9630825-11 1998 CONCLUSION: Heterozygotes for CYP2C9 I1e359/Leu allele have reduced in vivo metabolism of (S)-warfarin but not (R)-warfarin. Warfarin 90-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 9630825-12 1998 Because (S)-warfarin has a greater anticoagulant potency than its (R)-congener, the genetic polymorphism of CYP2C9 may partly account for the large interpatient variability in therapeutic dosages of warfarin. Warfarin 8-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 9630825-12 1998 Because (S)-warfarin has a greater anticoagulant potency than its (R)-congener, the genetic polymorphism of CYP2C9 may partly account for the large interpatient variability in therapeutic dosages of warfarin. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 9521735-3 1998 Sigmoidal kinetic characteristics were observed for carbamazepine metabolism by CYP3A4 and naphthalene metabolism by CYPs 2B6, 2C8, 2C9, and 3A5 as well as dapsone metabolism by CYP2C9. Carbamazepine 52-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 9531525-7 1998 Ketoconazole (5 microM), a selective CYP3A inhibitor, produced up to 75% inhibition, whereas other CYP-specific inhibitors, i.e. quinidine (CYP2D6), 7,8-benzoflavone (CYP1A2), and sulfaphenazole (CYP2C9), showed no significant effects. Ketoconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 9531513-7 1998 Microsomes from cells overexpressing CYP2D6 formed 5-HM, whereas N-dealkylated tolterodine was formed by microsomes expressing CYP2C9, -2C19, and -3A4. Tolterodine Tartrate 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 9531513-8 1998 The Km for formation of N-dealkylated tolterodine by CYP3A4 was similar to that obtained in human liver microsomes and higher for CYP2C9 and -2C19. Nitrogen 24-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-146 9531513-8 1998 The Km for formation of N-dealkylated tolterodine by CYP3A4 was similar to that obtained in human liver microsomes and higher for CYP2C9 and -2C19. Tolterodine Tartrate 38-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-146 9615477-1 1998 The 4"-hydroxylated metabolite of diclofenac was produced by biocatalysis for probing specific human drug-metabolising enzymes (CYP2C9). Diclofenac 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 9530332-5 1998 Deleted chromatin at the distal breakpoint includes the cytochrome P450IIC (CYP2C) gene cluster, thought to be involved in steroid hormone metabolism and therefore of possible significance to the growth rate of this androgen-dependent cell line. Steroids 123-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-81 9584328-7 1998 CYP1A1/2-, CYP2A6-, CYP2C9- and CYP2E1-associated activities were inhibited less potently or not at all by oxybutynin when compared with reference inhibitors. oxybutynin 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 9521735-3 1998 Sigmoidal kinetic characteristics were observed for carbamazepine metabolism by CYP3A4 and naphthalene metabolism by CYPs 2B6, 2C8, 2C9, and 3A5 as well as dapsone metabolism by CYP2C9. Dapsone 156-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 9521735-4 1998 Naphthalene metabolism by CYP3A4 and naproxen metabolism by CYP2C9 demonstrated nonhyperbolic enzyme kinetics suggestive of a low Km, low Vmax component for the first substrate molecule and a high Km, high Vmax component for the second substrate molecule. naphthalene 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 9521735-4 1998 Naphthalene metabolism by CYP3A4 and naproxen metabolism by CYP2C9 demonstrated nonhyperbolic enzyme kinetics suggestive of a low Km, low Vmax component for the first substrate molecule and a high Km, high Vmax component for the second substrate molecule. Naproxen 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 9521735-5 1998 7, 8-Benzoflavone activation of phenanthrene metabolism by CYP3A4 and dapsone activation of flurbiprofen and naproxen metabolism by CYP2C9 were also observed. alpha-naphthoflavone 0-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9521735-5 1998 7, 8-Benzoflavone activation of phenanthrene metabolism by CYP3A4 and dapsone activation of flurbiprofen and naproxen metabolism by CYP2C9 were also observed. phenanthrene 32-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9521735-5 1998 7, 8-Benzoflavone activation of phenanthrene metabolism by CYP3A4 and dapsone activation of flurbiprofen and naproxen metabolism by CYP2C9 were also observed. Dapsone 70-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9521735-5 1998 7, 8-Benzoflavone activation of phenanthrene metabolism by CYP3A4 and dapsone activation of flurbiprofen and naproxen metabolism by CYP2C9 were also observed. Flurbiprofen 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9521735-5 1998 7, 8-Benzoflavone activation of phenanthrene metabolism by CYP3A4 and dapsone activation of flurbiprofen and naproxen metabolism by CYP2C9 were also observed. Naproxen 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9534880-0 1998 Hepatic cytochrome P450 CYP2C activity in psoriasis: studies using proguanil as a probe compound. Proguanil 67-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-29 9586947-8 1998 Upon reconstitution, human liver b5 plus NADH:b5 reductase and CYP2C9 plus NADPH:P450 reductase were both effective catalysts of AZT reduction, which was also supported when CYP2A6 or CYP2E1 was substituted for CYP2C9. NAD 41-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 9586947-8 1998 Upon reconstitution, human liver b5 plus NADH:b5 reductase and CYP2C9 plus NADPH:P450 reductase were both effective catalysts of AZT reduction, which was also supported when CYP2A6 or CYP2E1 was substituted for CYP2C9. Zidovudine 129-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 9586947-8 1998 Upon reconstitution, human liver b5 plus NADH:b5 reductase and CYP2C9 plus NADPH:P450 reductase were both effective catalysts of AZT reduction, which was also supported when CYP2A6 or CYP2E1 was substituted for CYP2C9. Zidovudine 129-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 9534880-3 1998 We used proguanil as a probe to study CYP2C activity, and by implication retinol metabolism, in psoriasis. Proguanil 8-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-43 9542475-1 1998 BACKGROUND: Losartan is metabolized by CYP2C9 and CYP3A4 to an active metabolite, E3174, which has greater antihypertensive activity than the parent compound. Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 9549640-4 1998 Ritonavir had high inhibition potency against cytochrome P450-2C9 (tolbutamide hydroxylation), -2C19 (S-mephenytoin hydroxylation), and -2D6 (dextromethorphan O-demethylation and desipramine hydroxylation), while the other protease inhibitors had one or more orders of magnitude lower inhibitory activity against these reactions. Ritonavir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 9542475-1 1998 BACKGROUND: Losartan is metabolized by CYP2C9 and CYP3A4 to an active metabolite, E3174, which has greater antihypertensive activity than the parent compound. losartan carboxylic acid 82-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 9542475-13 1998 Further studies are needed to define the contribution of other isozymes, particularly CYP2C9, to the pharmacokinetics of losartan and E3174. Losartan 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 9542475-13 1998 Further studies are needed to define the contribution of other isozymes, particularly CYP2C9, to the pharmacokinetics of losartan and E3174. losartan carboxylic acid 134-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 9549641-7 1998 The CYP 1A2 inhibitor alpha-naphthoflavone and the CYP 2C9 inhibitor sulfaphenazole produced much less inhibition of amitriptyline N-demethylation at both substrate concentrations. Sulfaphenazole 69-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-58 9492390-0 1998 Cytochrome P450 2C9 catalyzes indomethacin O-demethylation in human liver microsomes. Indomethacin 30-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9492390-6 1998 When the capacity for indomethacin O-demethylation in microsomes of B lymphoblastoid cells expressing human CYPs was investigated at an indomethacin concentration of 5 microM, cDNA-expressed CYP2C9 exhibited 6-fold greater activity than did CYP2C19. Indomethacin 22-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 9492390-6 1998 When the capacity for indomethacin O-demethylation in microsomes of B lymphoblastoid cells expressing human CYPs was investigated at an indomethacin concentration of 5 microM, cDNA-expressed CYP2C9 exhibited 6-fold greater activity than did CYP2C19. Indomethacin 136-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 9492390-9 1998 Considering the 16-fold higher intrinsic clearance of CYP2C9, compared with that of CYP2C19, and these expression levels in human livers, the contribution of CYP2C19 to indomethacin O-demethylation was considered to be negligible. Indomethacin 169-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 9492390-10 1998 Indomethacin appears to be O-demethylated exclusively by CYP2C9 in humans. Indomethacin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 9491822-5 1998 RESULTS: Furafylline was a potent, selective inhibitor of phenacetin O-deethylation (CYP1A2-mediated) in human liver microsomes (IC50 = 0.48 microM), but inhibited both phenacetin O-deethylation and tolbutamide 4-hydroxylation (CYP2C9-mediated) at equimolar concentrations in rat liver microsomes (IC50 = 20.8 and 24.0 microM respectively). furafylline 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 228-234 9551703-1 1998 OBJECTIVE: Losartan is metabolised to its active metabolite E-3174 by CYP2C9 and CYP3A4 in vitro. Losartan 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 9551703-1 1998 OBJECTIVE: Losartan is metabolised to its active metabolite E-3174 by CYP2C9 and CYP3A4 in vitro. losartan carboxylic acid 60-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 9551703-2 1998 Itraconazole is an inhibitor of CYP3A4, whereas fluconazole affects CYP2C9 more than CYP3A4. Fluconazole 48-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 9551703-13 1998 This implicates that, in man, CYP2C9 is a major enzyme for the formation of E-3174 from losartan. losartan carboxylic acid 76-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 9551703-13 1998 This implicates that, in man, CYP2C9 is a major enzyme for the formation of E-3174 from losartan. Losartan 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Carbon-14 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9522436-5 1998 Humans with the Cys allele CYP2C9 variant, which was detected in 22% of Caucasians, were found to have similar catalytic rates to those of the wild-type in the oxidations of tolbutamide and racemic warfarin, whereas humans with the Leu allele, which was detected in 8% Japanese and 7% Caucasian samples, had lower catalytic rates than those of other two groups. Cysteine 16-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 9522436-5 1998 Humans with the Cys allele CYP2C9 variant, which was detected in 22% of Caucasians, were found to have similar catalytic rates to those of the wild-type in the oxidations of tolbutamide and racemic warfarin, whereas humans with the Leu allele, which was detected in 8% Japanese and 7% Caucasian samples, had lower catalytic rates than those of other two groups. Tolbutamide 174-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 9522436-5 1998 Humans with the Cys allele CYP2C9 variant, which was detected in 22% of Caucasians, were found to have similar catalytic rates to those of the wild-type in the oxidations of tolbutamide and racemic warfarin, whereas humans with the Leu allele, which was detected in 8% Japanese and 7% Caucasian samples, had lower catalytic rates than those of other two groups. Warfarin 198-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 9522436-5 1998 Humans with the Cys allele CYP2C9 variant, which was detected in 22% of Caucasians, were found to have similar catalytic rates to those of the wild-type in the oxidations of tolbutamide and racemic warfarin, whereas humans with the Leu allele, which was detected in 8% Japanese and 7% Caucasian samples, had lower catalytic rates than those of other two groups. Leucine 232-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 9522436-9 1998 Both human liver microsomes and recombinant CYP2C9 catalysed 7-hydroxylation of S-warfarin more extensively than those of R-warfarin. Warfarin 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 9522436-11 1998 Anti-CYP2C9 antibodies and sulphaphenazole inhibited the 6- and 7-hydroxylation of S-warfarin, but not R-warfarin, by > 90% and the methyl hydroxylation of tolbutamide by about 50%. Warfarin 83-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 9522436-11 1998 Anti-CYP2C9 antibodies and sulphaphenazole inhibited the 6- and 7-hydroxylation of S-warfarin, but not R-warfarin, by > 90% and the methyl hydroxylation of tolbutamide by about 50%. Tolbutamide 159-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Leucine 39-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Leucine 39-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Warfarin 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Warfarin 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-193 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Tolbutamide 113-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 9522436-13 1998 These results suggest that humans with Leu allele of CYP2C9 have lower Vmax"s for S-warfarin 7-hydroxylation and tolbutamide methyl hydroxylation than those with wild-type and Cys allele CYP2C9, although the K(m)"s are not very different in liver microsomes of these three groups of humans. Cysteine 176-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 9522436-14 1998 R-warfarin hydroxylation may be catalysed by P450 enzymes other than CYP2C9 in man. Warfarin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Carbon-14 54-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Linoleic Acid 58-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Carbon-14 54-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 11-hydroxyoctadecadienoic acid 87-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 11-hydroxy-9,12-octadecadienoic acid 119-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Arachidonic Acid 142-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. Carbon-14 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 13-hydroxyeicosatrienoic acid 210-239 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 13-Hete 241-248 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 10-hydroxyeicosatetraenoic acid 251-258 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-3 1998 CYP1A2, CYP2C8, CYP2C9, CYP2C19 and CYP3A4 converted [14C]linoleic acid to 14C-labeled 11-hydroxyoctadecadienoic acid (11-HODE), whereas [14C]arachidonic acid was oxygenated by CYP1A2 and CYP3A4 to 14C-labeled 13-hydroxyeicosatrienoic acid (13-HETE), 10-HETE and 7-HETE as determined by HPLC. 7-hydroxyeicosatetraenoic acid 263-269 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9435160-5 1998 CYP2C9 appeared to form 12R-HETE and 13-HETE, whereas CYP2C8 formed 13-HETE, 11-HETE and 15-HETE as main monohydroxy metabolites. 13-Hete 37-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9435160-5 1998 CYP2C9 appeared to form 12R-HETE and 13-HETE, whereas CYP2C8 formed 13-HETE, 11-HETE and 15-HETE as main monohydroxy metabolites. Hydroxyeicosatetraenoic Acids 27-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9416970-4 1997 Anti-CYP2C9 antibodies inhibited completely the 7-hydroxylation of S-warfarin, but not R-warfarin, catalyzed by human liver microsomes, while anti-CYP1A2 inhibited R-warfarin 7-hydroxylation by about 70%. Warfarin 67-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 9493314-8 1998 CYP2C9 and, to a much lesser extent, CYP3A4 were found to convert ME to M7. Meloxicam 66-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9416970-7 1997 These results suggest that R- and S-warfarin enantiomers are catalyzed principally by CYP1A2 and CYP2C9, respectively, in human liver microsomes, and that the pharmacokinetic properties of S-warfarin may be altered by R-warfarin in vivo when racemic warfarin is administered clinically to humans. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 14577221-0 1998 Determination of CYP2C9-catalyzed diclofenac 4"-hydroxylation by high-performance liquid chromatography. diclofenac 4" 34-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 9416970-5 1997 Interestingly, the racemic warfarin 7-hydroxylation activities (turnover numbers of 1.6 +/- 1.0 pmol/min/mg protein in 35 human samples) were found to be low compared with the S-warfarin 7-hydroxylation activities (4.1 +/- 2.5 pmol/min/mg protein), indicating that R-warfarin may have affected the CYP2C9-dependent S-warfarin 7-hydroxylation activities when racemic warfarin was used as a substrate. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 9416970-7 1997 These results suggest that R- and S-warfarin enantiomers are catalyzed principally by CYP1A2 and CYP2C9, respectively, in human liver microsomes, and that the pharmacokinetic properties of S-warfarin may be altered by R-warfarin in vivo when racemic warfarin is administered clinically to humans. Warfarin 34-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9416970-7 1997 These results suggest that R- and S-warfarin enantiomers are catalyzed principally by CYP1A2 and CYP2C9, respectively, in human liver microsomes, and that the pharmacokinetic properties of S-warfarin may be altered by R-warfarin in vivo when racemic warfarin is administered clinically to humans. Warfarin 218-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9431831-7 1997 Ketoconazole, an inhibitor of CYP3A4, inhibited competitively CPHP formation (Ki=0.1 microM), whereas sulphaphenazole (CYP2C9), furafylline (CYP1A2) and quinidine (CYP2D6) gave only little inhibition (IC50 > 100 microM). Sulfaphenazole 102-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 9431831-11 1997 HAL inhibited dextromethorphan O-demethylase (CYP2D6) with IC50 values between 2.7 and 8.5 microM, but not (IC50 > 100 microM) dextromethorphan N-demethylase (CYP3A4), phenacetin O-deethylase (CYP1A2) or tolbutamide hydroxylase (CYP2C9). Haloperidol 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 232-238 9384467-6 1997 Thus, assuming comparable molar concentrations at the site of inhibition, fluvoxamine can be expected to have the highest probability of interfering with the metabolism of CYP2C9 substrates. Fluvoxamine 74-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 9429229-3 1997 Similarly, the hydroxylation of tolbutamide, a marker for CYP2C9 was undetected in fetal liver microsomes and rose in the first month after birth. Tolbutamide 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 9429229-4 1997 In adult liver preparations, the hydroxylation of diazepam correlated well with the CYP3 A content of microsomes (r = 0.858, p < 0.01) and with the 6 beta hydroxylation of testosterone (r = 0.830, p < 0.005), whereas demethylation was related to the bulk of CYP2C proteins (r = 0.865, p < 0.005). Diazepam 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 264-269 9429229-7 1997 In newborns, barbiturates and to a lesser extent steroids, acted as inducers of CYP2C isoforms and increased tolbutamide hydroxylation, diazepam demethylation and diazepam hydroxylation by 2 to 10-fold. Barbiturates 13-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-85 9429229-7 1997 In newborns, barbiturates and to a lesser extent steroids, acted as inducers of CYP2C isoforms and increased tolbutamide hydroxylation, diazepam demethylation and diazepam hydroxylation by 2 to 10-fold. Steroids 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-85 9526963-0 1997 Warfarin analog inhibition of human CYP2C9-catalyzed S-warfarin 7-hydroxylation. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 9526963-0 1997 Warfarin analog inhibition of human CYP2C9-catalyzed S-warfarin 7-hydroxylation. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 9526963-1 1997 Human metabolism of the S-warfarin enantiomer is catalyzed primarily by cytochrome P4502C9 (CYP2C9), which, because of the enzyme"s broad drug substrate specificity, leads to drug-S-warfarin interactions. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 9526963-1 1997 Human metabolism of the S-warfarin enantiomer is catalyzed primarily by cytochrome P4502C9 (CYP2C9), which, because of the enzyme"s broad drug substrate specificity, leads to drug-S-warfarin interactions. Warfarin 182-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 9526963-2 1997 Several warfarin analogs have been synthesized and used to determine whether they exhibit diminished interactions with CYP2C9. Warfarin 8-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 9526963-3 1997 The kinetics of the warfarin analogs" inhibition of human liver microsomal CYP2C9 catalyzed metabolism of S-warfarin to S-7-hydroxywarfarin have been investigated. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 9526963-3 1997 The kinetics of the warfarin analogs" inhibition of human liver microsomal CYP2C9 catalyzed metabolism of S-warfarin to S-7-hydroxywarfarin have been investigated. Warfarin 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 9526963-3 1997 The kinetics of the warfarin analogs" inhibition of human liver microsomal CYP2C9 catalyzed metabolism of S-warfarin to S-7-hydroxywarfarin have been investigated. 7-hydroxywarfarin 120-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 9526963-6 1997 The fluorine substituted warfarins and the S-warfarin alcohol apparently bind with high affinity to CYP2C9. Fluorine 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9526963-6 1997 The fluorine substituted warfarins and the S-warfarin alcohol apparently bind with high affinity to CYP2C9. Warfarin 25-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9526963-6 1997 The fluorine substituted warfarins and the S-warfarin alcohol apparently bind with high affinity to CYP2C9. Sulfur 43-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9526963-6 1997 The fluorine substituted warfarins and the S-warfarin alcohol apparently bind with high affinity to CYP2C9. 6-hydroxywarfarin 45-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9526963-8 1997 The R-warfarin alcohol did not inhibit CYP2C9 catalyzed metabolism of S-warfarin and is less likely than warfarin to participate in CYP2C9 associated warfarin-drug interactions. r-warfarin alcohol 4-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9526963-8 1997 The R-warfarin alcohol did not inhibit CYP2C9 catalyzed metabolism of S-warfarin and is less likely than warfarin to participate in CYP2C9 associated warfarin-drug interactions. Warfarin 6-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 9384467-7 1997 S-fluoxetine is on average a 5 fold weaker CYP2C9 inhibitor than either R-fluoxetine or the racemic mixture. Fluoxetine 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 9353388-0 1997 Human CYP2C9 and CYP2A6 mediate formation of the hepatotoxin 4-ene-valproic acid. 2-propyl-4-pentenoic acid 61-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 9390115-5 1997 The influence of ticlopidine on CYP2C9, the other cytochrome P450 isoform that metabolizes phenytoin, is relatively weak, with a calculated Ki of 38.8 +/- 27 mumol/L. Ticlopidine 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9390115-5 1997 The influence of ticlopidine on CYP2C9, the other cytochrome P450 isoform that metabolizes phenytoin, is relatively weak, with a calculated Ki of 38.8 +/- 27 mumol/L. Phenytoin 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Omeprazole 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 336-342 9353355-5 1997 Omeprazole 5-hydroxylation by liver microsomes of a human sample that contained relatively high levels of CYP3A4 and low levels of CYP2C19 were inhibited very significantly by ketoconazole and anti-CYP3A4 antibodies, although a human sample having high in CYP2C19 and low in CYP3A4 was found to be sensitive toward fluvoxamine and anti-CYP2C9 antibodies. Ketoconazole 176-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 336-342 9335524-1 1997 Purified recombinant human liver cytochrome P450 2C9 was produced, from expression of the corresponding cDNA in yeast, in quantities large enough for UV-visible and 1H NMR experiments. Hydrogen 165-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-52 9335524-4 1997 On the contrary, sulfaphenazole led to a low-spin (S = 1/2) CYP 2C9 complex upon binding of its NH2 group to CYP 2C9 iron. Sulfaphenazole 17-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-67 9335524-4 1997 On the contrary, sulfaphenazole led to a low-spin (S = 1/2) CYP 2C9 complex upon binding of its NH2 group to CYP 2C9 iron. Sulfaphenazole 17-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-116 9335524-4 1997 On the contrary, sulfaphenazole led to a low-spin (S = 1/2) CYP 2C9 complex upon binding of its NH2 group to CYP 2C9 iron. Iron 117-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-67 9335524-4 1997 On the contrary, sulfaphenazole led to a low-spin (S = 1/2) CYP 2C9 complex upon binding of its NH2 group to CYP 2C9 iron. Iron 117-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-116 9335524-5 1997 Interactions of the five substrates with the enzyme were studied by paramagnetic relaxation effects of CYP 2C9-iron(III) on the 1H NMR spectrum of each substrate. Iron 111-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-110 9335524-5 1997 Interactions of the five substrates with the enzyme were studied by paramagnetic relaxation effects of CYP 2C9-iron(III) on the 1H NMR spectrum of each substrate. Hydrogen 128-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-110 9335524-7 1997 Finally, a model for substrate positioning in the CYP 2C9 active site was constructed by molecular modeling studies under the constraint of the iron-proton distances. Iron 144-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-57 9335524-9 1997 Sulfaphenazole was easily included in that model; its very high affinity for CYP 2C9 is due to a third structural feature, the presence of its NH2 function which binds to CYP 2C9 iron. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-84 9335524-9 1997 Sulfaphenazole was easily included in that model; its very high affinity for CYP 2C9 is due to a third structural feature, the presence of its NH2 function which binds to CYP 2C9 iron. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-178 9335524-9 1997 Sulfaphenazole was easily included in that model; its very high affinity for CYP 2C9 is due to a third structural feature, the presence of its NH2 function which binds to CYP 2C9 iron. Iron 179-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-84 9335524-9 1997 Sulfaphenazole was easily included in that model; its very high affinity for CYP 2C9 is due to a third structural feature, the presence of its NH2 function which binds to CYP 2C9 iron. Iron 179-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-178 9328296-3 1997 These steroid chemicals also inhibited CYP2C9-dependent S-warfarin 7-hydroxylation activities though lesser extents seen with those in CYP2C19 enzyme. Steroids 6-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 9328296-3 1997 These steroid chemicals also inhibited CYP2C9-dependent S-warfarin 7-hydroxylation activities though lesser extents seen with those in CYP2C19 enzyme. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 9328296-4 1997 Progesterone was found to be a competitive inhibitor of CYP2C19 and CYP2C9 in human liver microsomes. Progesterone 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 9328296-6 1997 CYP2C9 also had progesterone 21-hydroxylation activities, although the activities were lower than those catalyzed by CYP2C19. Progesterone 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9328296-9 1997 Immunoinhibition experiments suggested that anti-CYP2C9 (which inhibits both CYP2C9 and CYP2C19 catalytic activities) suppressed the progesterone 21-hydroxylation activities catalyzed by liver microsomes of humans and monkeys and that anti-CYP2C11 inhibited the progesterone 21-hydroxylation activities catalyzed by liver microsomes of male rats. Progesterone 133-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 9328296-9 1997 Immunoinhibition experiments suggested that anti-CYP2C9 (which inhibits both CYP2C9 and CYP2C19 catalytic activities) suppressed the progesterone 21-hydroxylation activities catalyzed by liver microsomes of humans and monkeys and that anti-CYP2C11 inhibited the progesterone 21-hydroxylation activities catalyzed by liver microsomes of male rats. Progesterone 133-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 9328296-9 1997 Immunoinhibition experiments suggested that anti-CYP2C9 (which inhibits both CYP2C9 and CYP2C19 catalytic activities) suppressed the progesterone 21-hydroxylation activities catalyzed by liver microsomes of humans and monkeys and that anti-CYP2C11 inhibited the progesterone 21-hydroxylation activities catalyzed by liver microsomes of male rats. Progesterone 262-274 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 9328296-9 1997 Immunoinhibition experiments suggested that anti-CYP2C9 (which inhibits both CYP2C9 and CYP2C19 catalytic activities) suppressed the progesterone 21-hydroxylation activities catalyzed by liver microsomes of humans and monkeys and that anti-CYP2C11 inhibited the progesterone 21-hydroxylation activities catalyzed by liver microsomes of male rats. Progesterone 262-274 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 9328296-11 1997 Androstenedione formation catalyzed by liver microsomes of humans and monkeys and of male rats was suppressed by anti-CYP2C9 and anti-CYP2C11, respectively. Androstenedione 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 9328296-13 1997 CYP2C9 may have some, but lesser extent than those by CYP2C19, of the catalytic roles for the metabolism of progesterone and testosterone by human liver microsomes. Progesterone 108-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9328296-13 1997 CYP2C9 may have some, but lesser extent than those by CYP2C19, of the catalytic roles for the metabolism of progesterone and testosterone by human liver microsomes. Testosterone 125-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 9352571-0 1997 Genetic association between sensitivity to warfarin and expression of CYP2C9*3. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 9331082-6 1997 On the basis of cytochrome P450 (CYP) isoform-specific chemical inhibitor and cDNA-expressed human P450 isozyme studies, CYP2C9 and CYP3A4/5 seemed to be the major P450 isoforms responsible for HCY formation at low (0.1 mM) and high (0.7 and 5 mM) concentrations of CY, respectively. 4-hydroxycyclophosphamide 194-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 9331082-6 1997 On the basis of cytochrome P450 (CYP) isoform-specific chemical inhibitor and cDNA-expressed human P450 isozyme studies, CYP2C9 and CYP3A4/5 seemed to be the major P450 isoforms responsible for HCY formation at low (0.1 mM) and high (0.7 and 5 mM) concentrations of CY, respectively. Cyclophosphamide 33-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 9352571-1 1997 Cytochrome P4502C9 (CYP2C9) is largely responsible for terminating the anticoagulant effect of racemic warfarin via hydroxylation of the pharmacologically more potent S-enantiomer to inactive metabolites. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-18 9352571-1 1997 Cytochrome P4502C9 (CYP2C9) is largely responsible for terminating the anticoagulant effect of racemic warfarin via hydroxylation of the pharmacologically more potent S-enantiomer to inactive metabolites. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 9352571-9 1997 Therefore, expression of CYP2C9*3 is associated with diminished clearance of S-warfarin and a dangerously exacerbated therapeutic response to normal doses of the racemic drug. Sulfur 77-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 9352571-9 1997 Therefore, expression of CYP2C9*3 is associated with diminished clearance of S-warfarin and a dangerously exacerbated therapeutic response to normal doses of the racemic drug. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 9298519-8 1997 Cytochrome P450-2C9 appears to be the principal human cytochrome mediating fluoxetine N-demethylation. Fluoxetine 75-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 9333104-1 1997 OBJECTIVE: To examine the genetic polymorphism of CYP2C9 and CYP2C19 and its effect on the pharmacokinetics of phenytoin among 44 Japanese patients with epilepsy. Phenytoin 111-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 9278209-11 1997 Saquinavir was the least potent CYP3A4 inhibitor (K(i) = 2.99 +/- 0.87 microM) and produced some inhibition of CYP2C9 (approximately 50% at 50 microM). Saquinavir 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Saquinavir 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Ritonavir 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 9278209-1 1997 AIMS: To compare the inhibitory potential of the HIV protease inhibitors saquinavir, ritonavir and indinavir against CYP1A2, CYP2C9, CYP2E1 and CYP3A4 catalysed metabolic reactions in human liver microsomes in vitro. Indinavir 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 9205822-0 1997 Venlafaxine: in vitro inhibition of CYP2D6 dependent imipramine and desipramine metabolism; comparative studies with selected SSRIs, and effects on human hepatic CYP3A4, CYP2C9 and CYP1A2. Venlafaxine Hydrochloride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Tolbutamide 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). 4'-hydroxytolbutamide 88-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Omeprazole 142-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Lansoprazole 185-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Dextromethorphan 267-283 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9296349-5 1997 cDNA-expressed CYP2C9 (Arg 144 and Cys 144) favored S-2- and S-3-hydroxyibuprofen formation, but CYP2C8 favored R-2-hydroxyibuprofen formation. Arginine 23-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9296349-5 1997 cDNA-expressed CYP2C9 (Arg 144 and Cys 144) favored S-2- and S-3-hydroxyibuprofen formation, but CYP2C8 favored R-2-hydroxyibuprofen formation. Cysteine 35-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9296349-5 1997 cDNA-expressed CYP2C9 (Arg 144 and Cys 144) favored S-2- and S-3-hydroxyibuprofen formation, but CYP2C8 favored R-2-hydroxyibuprofen formation. s-2- and s-3-hydroxyibuprofen 52-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9296349-10 1997 The relative levels of both CYP2C8 and CYP2C9 and the expression of the corresponding variants may influence the disposition of ibuprofen in vivo. Ibuprofen 128-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). 3-methoxymorphinan 287-305 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Omeprazole 338-348 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9224780-6 1997 Both drugs were competitive inhibitors of CYP2C9-catalyzed conversion of tolbutamide to 4-hydroxytolbutamide (K(i) = 40.1 +/- 14.8 microM for omeprazole, K(i) = 52.1 +/- 1.4 microM for lansoprazole) and were noncompetitive inhibitors of CYP3A-catalyzed conversion of dextromethorphan to 3-methoxymorphinan (K(i) = 84.4 +/- 4.0 microM for omeprazole, K(i) = 170.4 +/- 7.1 microM for lansoprazole). Lansoprazole 382-394 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 9223567-7 1997 CYP3A4 in lymphoblastoid cell microsomes catalyzed DTZ N-demethylation but CYP2C8 and CYP2C9 were also active (approximately 20% and 10% of the activity supported by CYP3A4); seven other CYPs produced little or no N-desmethyl DTZ from DTZ. N-monodemethyldiltiazem 214-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 9186495-2 1997 CYP2C10 required cytochrome b5 (b5) for optimal rates of tolbutamide and S-warfarin oxidations and b5 could be replaced by apo-b5; apo-b5 and b5 effects on the reconstituted systems have already been reported in systems containing CYP3A4 for the oxidation of testosterone and nifedipine and for the rapid reduction of CYP3A4 by NADPH-P450 reductase (H. Yamazaki et al., 1996, J. Biol. Tolbutamide 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 9186495-2 1997 CYP2C10 required cytochrome b5 (b5) for optimal rates of tolbutamide and S-warfarin oxidations and b5 could be replaced by apo-b5; apo-b5 and b5 effects on the reconstituted systems have already been reported in systems containing CYP3A4 for the oxidation of testosterone and nifedipine and for the rapid reduction of CYP3A4 by NADPH-P450 reductase (H. Yamazaki et al., 1996, J. Biol. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 9186495-2 1997 CYP2C10 required cytochrome b5 (b5) for optimal rates of tolbutamide and S-warfarin oxidations and b5 could be replaced by apo-b5; apo-b5 and b5 effects on the reconstituted systems have already been reported in systems containing CYP3A4 for the oxidation of testosterone and nifedipine and for the rapid reduction of CYP3A4 by NADPH-P450 reductase (H. Yamazaki et al., 1996, J. Biol. Testosterone 259-271 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 9186495-2 1997 CYP2C10 required cytochrome b5 (b5) for optimal rates of tolbutamide and S-warfarin oxidations and b5 could be replaced by apo-b5; apo-b5 and b5 effects on the reconstituted systems have already been reported in systems containing CYP3A4 for the oxidation of testosterone and nifedipine and for the rapid reduction of CYP3A4 by NADPH-P450 reductase (H. Yamazaki et al., 1996, J. Biol. Nifedipine 276-286 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 9186495-8 1997 Interestingly, we found that testosterone 6 beta-hydroxylation by CYP3A4 was stimulated by CYP1A2 (and also by a modified form in which the first 36 residues of the native human protein were removed) and CYP1A1 as well as by b5, and such stimulations were not seen when other P450 proteins (e.g., CYP2C10, 2D6, or 2E1) were added to the reconstituted systems. Testosterone 29-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 297-304 9205822-6 1997 Venlafaxine"s IC50 values for CYP3A4, CYP1A2 CYP2C9 were determined based on inhibition of probe substrate activities (testosterone 6 beta-hydroxylation, ethoxyresorufin O-dealkylase and tolbutamide 4-hydroxylation, respectively). Venlafaxine Hydrochloride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 9241661-11 1997 Thus (a) wild type CYP2C19 and CYP2C9 are relatively low Km catalysts of cyclophosphamide and ifosfamide activation, and (b) all four human CYP2C enzymes activate these two anticancer prodrugs with varying efficiencies and with striking differences among naturally occurring allelic variants in the case of CYP2C9 and CYP2C18. Ifosfamide 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 307-313 9241660-6 1997 The rates of (S)-warfarin 7-hydroxylation by CYP2C9 and the R144C variant also exhibited differential dependence on salt concentration which further supported a difference in interaction with OR. Warfarin 13-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 9157990-4 1997 These inductions were associated with corresponding increases in immunoreactive CYP2B6, CYP2C8, CYP2C9, and CYP3A4, all previously shown to catalyze oxazaphosphorine activation. oxazaphosphorine 149-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 9241660-6 1997 The rates of (S)-warfarin 7-hydroxylation by CYP2C9 and the R144C variant also exhibited differential dependence on salt concentration which further supported a difference in interaction with OR. Salts 116-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 9241661-0 1997 Identification of the polymorphically expressed CYP2C19 and the wild-type CYP2C9-ILE359 allele as low-Km catalysts of cyclophosphamide and ifosfamide activation. Cyclophosphamide 118-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 9241661-0 1997 Identification of the polymorphically expressed CYP2C19 and the wild-type CYP2C9-ILE359 allele as low-Km catalysts of cyclophosphamide and ifosfamide activation. Ifosfamide 139-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 9241661-3 1997 Recombinant CYP2C8, CYP2C19, two allelic variants of CYP2C18, and six variants of CYP2C9 expressed in a yeast cDNA expression system were each enzymatically active, as judged by the ability of the isolated microsomes to catalyse 7-ethoxycoumarin O-deethylation after reconstitution with purified NADPH-cytochrome P450 reductase and cytochrome b5. 3-ethoxychromen-2-one 231-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 9241661-4 1997 With cyclophosphamide as substrate, CYP2C19 had the lowest apparent Km, followed by CYP2C9, CYP2C18 and CYP2C8, whereas in the case of ifosfamide, the rank order was: Km CYP2C19 < CYP2C18 < CYP2C9 < CYP2C8. Cyclophosphamide 5-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 9241661-4 1997 With cyclophosphamide as substrate, CYP2C19 had the lowest apparent Km, followed by CYP2C9, CYP2C18 and CYP2C8, whereas in the case of ifosfamide, the rank order was: Km CYP2C19 < CYP2C18 < CYP2C9 < CYP2C8. Cyclophosphamide 5-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 9241661-4 1997 With cyclophosphamide as substrate, CYP2C19 had the lowest apparent Km, followed by CYP2C9, CYP2C18 and CYP2C8, whereas in the case of ifosfamide, the rank order was: Km CYP2C19 < CYP2C18 < CYP2C9 < CYP2C8. Ifosfamide 135-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 9241661-7 1997 In the case of CYP2C9, a Ile359 to Leu mutation associated with poor metabolism of the hypoglycemic drug tolbutamide decreased catalytic efficiency toward cyclophosphamide by increasing the apparent Km, whereas the same mutation reduced the efficiency of this P450 toward ifosfamide by decreasing the Vmax. Tolbutamide 105-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9241661-7 1997 In the case of CYP2C9, a Ile359 to Leu mutation associated with poor metabolism of the hypoglycemic drug tolbutamide decreased catalytic efficiency toward cyclophosphamide by increasing the apparent Km, whereas the same mutation reduced the efficiency of this P450 toward ifosfamide by decreasing the Vmax. Cyclophosphamide 155-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9241661-7 1997 In the case of CYP2C9, a Ile359 to Leu mutation associated with poor metabolism of the hypoglycemic drug tolbutamide decreased catalytic efficiency toward cyclophosphamide by increasing the apparent Km, whereas the same mutation reduced the efficiency of this P450 toward ifosfamide by decreasing the Vmax. Ifosfamide 272-282 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 9241661-8 1997 Substitution of CYP2C9-Gly417 by Asp resulted in a two-fold lower catalytic efficiency for cyclophosphamide metabolism but a three-fold higher efficiency for ifosfamide metabolism. Cyclophosphamide 91-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9241661-8 1997 Substitution of CYP2C9-Gly417 by Asp resulted in a two-fold lower catalytic efficiency for cyclophosphamide metabolism but a three-fold higher efficiency for ifosfamide metabolism. Ifosfamide 158-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 9241661-11 1997 Thus (a) wild type CYP2C19 and CYP2C9 are relatively low Km catalysts of cyclophosphamide and ifosfamide activation, and (b) all four human CYP2C enzymes activate these two anticancer prodrugs with varying efficiencies and with striking differences among naturally occurring allelic variants in the case of CYP2C9 and CYP2C18. Cyclophosphamide 73-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 9241661-11 1997 Thus (a) wild type CYP2C19 and CYP2C9 are relatively low Km catalysts of cyclophosphamide and ifosfamide activation, and (b) all four human CYP2C enzymes activate these two anticancer prodrugs with varying efficiencies and with striking differences among naturally occurring allelic variants in the case of CYP2C9 and CYP2C18. Cyclophosphamide 73-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 307-313 9241661-11 1997 Thus (a) wild type CYP2C19 and CYP2C9 are relatively low Km catalysts of cyclophosphamide and ifosfamide activation, and (b) all four human CYP2C enzymes activate these two anticancer prodrugs with varying efficiencies and with striking differences among naturally occurring allelic variants in the case of CYP2C9 and CYP2C18. Ifosfamide 94-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 9157990-6 1997 CYP3A4, CYP2C8, and CYP2C9 protein levels were also increased by exposure of the hepatocytes to cyclophosphamide or ifosfamide (50 microM), which thereby enhanced their own rates of 4-hydroxylation in the cultured hepatocytes. Cyclophosphamide 96-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 9157990-6 1997 CYP3A4, CYP2C8, and CYP2C9 protein levels were also increased by exposure of the hepatocytes to cyclophosphamide or ifosfamide (50 microM), which thereby enhanced their own rates of 4-hydroxylation in the cultured hepatocytes. Ifosfamide 116-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 9089421-3 1997 Incubation of a fixed concentration of mestranol (3 mumol/L) with varying concentrations of CYP inhibitors revealed strong inhibition of ethinyl estradiol formation by sulfaphenazole, a specific CYP2C9 inhibitor, with an average inhibitor concentration at one half of Emax (IC50) of 3.6 mumol/L (range, 1.8-8.3 mumol/L) and an average maximal inhibitory capacity (Emax) of 75% (range, 60-91%). Mestranol 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 9089421-8 1997 Strong inhibition of ethinyl estradiol formation by sulfaphenazole suggests a major contribution of CYP2C9 to this reaction. Ethinyl Estradiol 21-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9115051-14 1997 Clearances of the two NSAIDs were significantly correlated, suggesting that a common P450 isozyme (most likely CYP2C9, in that piroxicam is a known substrate of this isozyme) may be at least partly involved in the oxidative metabolism of these NSAIDs. Piroxicam 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 9170147-0 1997 Relationship between CYP2C9 and 2C19 genotypes and tolbutamide methyl hydroxylation and S-mephenytoin 4"-hydroxylation activities in livers of Japanese and Caucasian populations. Tolbutamide 51-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 9170147-10 1997 Good correlations were found between levels of CYP2C9 and activities of tolbutamide methyl hydroxylation (r = 0.77) and between levels of CYP2C19 and activities of S-mephenytoin 4"-hydroxylation (r = 0.86) in liver microsomes of the human samples examined. Tolbutamide 72-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 9170147-11 1997 Tolbutamide methyl hydroxylation activities were lower in human samples with the Leu359 allele of CYP2C9 than those with the Cys144 allele and wild-type (Arg144-Ile359); the former type showed slightly higher K(m) values. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 9172960-6 1997 In human liver microsomes, orphenadrine strongly decreased the marker activities of CYP2B6, CYP2D6, as well as CYP2C9; and partially decreased the marker activities of CYP1A2, CYP2A6, CYP3A4, and CYP2C19. Orphenadrine 27-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 9172960-7 1997 In the competitive inhibition protocol, methimazole had no effect on the marker activities of CYP2E1 and CYP2A6; slightly decreased CYP2D6 marker activity; partially decreased the marker activities of CYP2C19, CYP2C9, and CYP2B6; and dramatically decreased CYP3A4 marker activity. Methimazole 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 210-216 9089421-8 1997 Strong inhibition of ethinyl estradiol formation by sulfaphenazole suggests a major contribution of CYP2C9 to this reaction. Sulfaphenazole 52-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 9089421-3 1997 Incubation of a fixed concentration of mestranol (3 mumol/L) with varying concentrations of CYP inhibitors revealed strong inhibition of ethinyl estradiol formation by sulfaphenazole, a specific CYP2C9 inhibitor, with an average inhibitor concentration at one half of Emax (IC50) of 3.6 mumol/L (range, 1.8-8.3 mumol/L) and an average maximal inhibitory capacity (Emax) of 75% (range, 60-91%). Ethinyl Estradiol 137-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 9089421-3 1997 Incubation of a fixed concentration of mestranol (3 mumol/L) with varying concentrations of CYP inhibitors revealed strong inhibition of ethinyl estradiol formation by sulfaphenazole, a specific CYP2C9 inhibitor, with an average inhibitor concentration at one half of Emax (IC50) of 3.6 mumol/L (range, 1.8-8.3 mumol/L) and an average maximal inhibitory capacity (Emax) of 75% (range, 60-91%). Sulfaphenazole 168-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 9089421-5 1997 alpha-Naphthoflavone (a CYP1A1/2 inhibitor only at concentrations < 2 mumol/L and a CYP2C9 inhibitor at higher concentrations) had a weak inhibitory effect on ethinyl estradiol formation (< 20% decrease in mestranol demethylation activity). alpha-naphthoflavone 0-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 9089421-5 1997 alpha-Naphthoflavone (a CYP1A1/2 inhibitor only at concentrations < 2 mumol/L and a CYP2C9 inhibitor at higher concentrations) had a weak inhibitory effect on ethinyl estradiol formation (< 20% decrease in mestranol demethylation activity). Ethinyl Estradiol 162-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 9131945-5 1997 A further decrease in activity was associated with the addition of the CYP2C9 inhibitor sulphaphenazole such that the residual kinetics were best described by a single Michaelis-Menten function. Sulfaphenazole 88-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 9131945-9 1997 Microsomes from yeast cells expressing CYP2D6 and from human lymphoblastoid cells expressing CYP3A4 or CYP2C9-Arg N-demethylated AMI, but those from cells expressing CYPs 1A2 and 2C19 did not. Arginine 110-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 9131945-9 1997 Microsomes from yeast cells expressing CYP2D6 and from human lymphoblastoid cells expressing CYP3A4 or CYP2C9-Arg N-demethylated AMI, but those from cells expressing CYPs 1A2 and 2C19 did not. Nitrogen 114-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 9131945-9 1997 Microsomes from yeast cells expressing CYP2D6 and from human lymphoblastoid cells expressing CYP3A4 or CYP2C9-Arg N-demethylated AMI, but those from cells expressing CYPs 1A2 and 2C19 did not. Amitriptyline 129-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 9068931-22 1997 Fluoxetine substantially inhibits CYP2D6 and probably CYP2C9/10, moderately inhibits CYP2C19 and mildly inhibits CYP3A3/4. Fluoxetine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 9029042-6 1997 At a substrate concentration of 10 microM, diazepam N-demethylation in a panel of human liver microsomes was inhibited 42 +/- 12% (mean +/- SD, N = 6) by a polyclonal anti-CYP2C antibody. Diazepam 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-177 9023313-12 1997 Testosterone 6beta-hydroxylation (mediated by CYP3A4), 7-ethylresorufin O-deethylation (CYP1A2) and tolbutamide methyl hydroxylation (CYP2C9/10), but not aniline 4-hydroxylation (CYP2E1), were inhibited effectively by parathion. Testosterone 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 9023313-12 1997 Testosterone 6beta-hydroxylation (mediated by CYP3A4), 7-ethylresorufin O-deethylation (CYP1A2) and tolbutamide methyl hydroxylation (CYP2C9/10), but not aniline 4-hydroxylation (CYP2E1), were inhibited effectively by parathion. aniline 154-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 9023313-12 1997 Testosterone 6beta-hydroxylation (mediated by CYP3A4), 7-ethylresorufin O-deethylation (CYP1A2) and tolbutamide methyl hydroxylation (CYP2C9/10), but not aniline 4-hydroxylation (CYP2E1), were inhibited effectively by parathion. Parathion 218-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 9037249-6 1997 Inhibition of activity by quinidine (1 microM), sulphaphenazole (20 microM) and ketoconazole (2 microM), selective inhibitors of CYPs 2D6, 2C9 and 3A4, respectively, was 0-80%, 0-80% and 12-57%. Quinidine 26-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-142 9037249-6 1997 Inhibition of activity by quinidine (1 microM), sulphaphenazole (20 microM) and ketoconazole (2 microM), selective inhibitors of CYPs 2D6, 2C9 and 3A4, respectively, was 0-80%, 0-80% and 12-57%. Sulfaphenazole 48-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-142 9037249-6 1997 Inhibition of activity by quinidine (1 microM), sulphaphenazole (20 microM) and ketoconazole (2 microM), selective inhibitors of CYPs 2D6, 2C9 and 3A4, respectively, was 0-80%, 0-80% and 12-57%. Ketoconazole 80-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-142 8960070-7 1997 The formation rates of both colchicine metabolites were correlated significantly with nifedipine oxidase activity, a marker of CYP3A4 activity (r = 0.96, P < 0.001), but not with the metabolic markers of CYP2A6, CYP2C19, CYP2C9, CYP2D6, and CYP2E1 activities. Colchicine 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-230 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Tolbutamide 33-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 9068933-5 1997 This suggests that the change in tolbutamide clearance may be due to a slight inhibition of the cytochrome P450 (CYP) isoenzyme CYP2C9/10 when sertraline was administered in its maximum recommended dosage. Sertraline 143-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 9068934-8 1997 The metabolism of warfarin is principally mediated by the cytochrome P450 (CYP) isoenzyme CYP2C9/10. Warfarin 18-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Arginine 151-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Arginine 151-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Cysteine 155-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Cysteine 155-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Tyrosine 173-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Tyrosine 173-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Cysteine 177-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Cysteine 177-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Isoleucine 195-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Isoleucine 195-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Leucine 199-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Leucine 199-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Glycine 220-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Glycine 220-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Aspartic Acid 224-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 9110362-1 1997 Single nucleotide substitutions are known to result in a different amino acid at one of four sites in cytochrome P4502C9 (CYP2C9) namely: residue 144: Arg/Cys; residue 358: Tyr/Cys; residue 359: Ile/Leu and residue 417: Gly/Asp. Aspartic Acid 224-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 9010637-5 1997 The inducers of CYP3A and/or CYP2C9 (e.g. rifampicin and phenytoin) are likely to alter the disposition of seratrodast. Rifampin 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 9010637-5 1997 The inducers of CYP3A and/or CYP2C9 (e.g. rifampicin and phenytoin) are likely to alter the disposition of seratrodast. Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 9021169-1 1996 The metabolism of isoprene was investigated with microsomes derived from cell lines expressing human CYP1A1, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, CYP2E1, or CYP3A4. isoprene 18-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 8973193-0 1996 Interaction of sulfaphenazole derivatives with human liver cytochromes P450 2C: molecular origin of the specific inhibitory effects of sulfaphenazole on CYP 2C9 and consequences for the substrate binding site topology of CYP 2C9. Sulfaphenazole 15-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-160 8973193-0 1996 Interaction of sulfaphenazole derivatives with human liver cytochromes P450 2C: molecular origin of the specific inhibitory effects of sulfaphenazole on CYP 2C9 and consequences for the substrate binding site topology of CYP 2C9. Sulfaphenazole 15-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-228 8973193-0 1996 Interaction of sulfaphenazole derivatives with human liver cytochromes P450 2C: molecular origin of the specific inhibitory effects of sulfaphenazole on CYP 2C9 and consequences for the substrate binding site topology of CYP 2C9. Sulfaphenazole 135-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-160 8973193-3 1996 From difference visible spectroscopy experiments using microsomes of yeast expressing various human P450s, 1 selectively interacts only with CYP 2C9 with the appearance of a peak at 429 nm as expected for the formation of a P450 Fe(III)-nitrogenous ligand complex (Ks = 0.4 +/- 0.1 microM). ferric sulfate 229-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-148 8973193-3 1996 From difference visible spectroscopy experiments using microsomes of yeast expressing various human P450s, 1 selectively interacts only with CYP 2C9 with the appearance of a peak at 429 nm as expected for the formation of a P450 Fe(III)-nitrogenous ligand complex (Ks = 0.4 +/- 0.1 microM). Potassium 265-267 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-148 8973193-4 1996 Comparative studies of the spectral interaction and inhibitory effects of twelve compounds related to 1 with CYP 2C9 showed that the aniline function of 1 is responsible for the formation of the iron-nitrogen bond of the 429 nm-absorbing complex and is necessary for the inhibitory effects of 1. aniline 133-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-116 8973193-4 1996 Comparative studies of the spectral interaction and inhibitory effects of twelve compounds related to 1 with CYP 2C9 showed that the aniline function of 1 is responsible for the formation of the iron-nitrogen bond of the 429 nm-absorbing complex and is necessary for the inhibitory effects of 1. Iron 195-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-116 8973193-4 1996 Comparative studies of the spectral interaction and inhibitory effects of twelve compounds related to 1 with CYP 2C9 showed that the aniline function of 1 is responsible for the formation of the iron-nitrogen bond of the 429 nm-absorbing complex and is necessary for the inhibitory effects of 1. Nitrogen 200-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-116 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Nitrogen 244-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-43 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Nitrogen 244-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Nitrogen 244-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Nitrogen 244-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Iron 269-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-43 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Iron 269-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Iron 269-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. Iron 269-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. so2n 308-312 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-43 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. so2n 308-312 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. so2n 308-312 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8973193-6 1996 A model for the binding of 1 in the CYP 2C9 active site is proposed; that takes into account three major interactions that should be at the origin of the high-affinity and specific inhibitory effects of 1 toward CYP 2C9: (i) the binding of its nitrogen atom to CYP 2C9 iron, (ii) an ionic interaction of its SO2N- anionic site with a cationic residue of CYP 2C9, and (iii) an interaction of its N-phenyl group with an hydrophobic part of the protein active site. so2n 308-312 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-219 8944768-8 1996 Mechanism-based inactivation of cytochrome P-450 during tienilic acid oxidation was observed in the case of CYP 2C9 but was not detectable with CYP 2C18 and CYP 2C8. Ticrynafen 56-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-115 9004453-3 1996 A retroviral vector construct, containing a human CYP2C10 cDNA, was transfected in V79-NH Chinese hamster lung cells by calcium phosphate co-precipitation. calcium phosphate 120-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-57 9004453-6 1996 Enzymatic activity of CYP2C10 was detected by 4-methylhydroxylation of tolbutamide. Tolbutamide 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-29 9032002-6 1996 Fluoxetine is a substantial inhibitor of CYP2D6, has mild effects on CYP3A3/4, and may also have effects on CYP2C9/10 and CYP2C19. Fluoxetine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 8944768-2 1996 Oxidation of tienilic acid by human cytochromes P-450 (CYP) 2C9, 2C18, 2C8 and 2C19 was studied using recombinant enzymes expressed in yeast. Ticrynafen 13-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-63 8944768-9 1996 Tienilic acid thus appears to be a mechanism-based inhibitor specific for CYP 2C9 in human liver. Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-81 8944768-3 1996 CYP 2C9 was the best catalyst for 5-hydroxylation of tienilic acid (K(m) = 5 +/- 1 microM, kcat = 1.7 +/- 0.2 min-1), 30-fold more potent in terms of kcat/K(m) than CYP 2C18 (K(m) = 150 +/- 15 microM, kcat = 1.8 +/- 0.2 min-1) and 300-fold more potent than CYP 2C8 (K(m) = 145 +/- 15 microM, kcat = 0.2 +/- 0.1 min-1). Ticrynafen 53-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 8894508-7 1996 CYP2C19, CYP2C8, CYP2C18, and CYP2C9 formed a single metabolite with an HPLC retention time identical to that of 5OH-OP. 5oh 113-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 8944768-7 1996 Specific-covalent binding of tienilic acid metabolites to cytochrome P-450 (incubations in the presence of 5 mM glutathione) was markedly higher upon tienilic acid oxidation by CYP 2C9 than by CYP 2C18 and CYP 2C8. Ticrynafen 29-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-184 8944768-7 1996 Specific-covalent binding of tienilic acid metabolites to cytochrome P-450 (incubations in the presence of 5 mM glutathione) was markedly higher upon tienilic acid oxidation by CYP 2C9 than by CYP 2C18 and CYP 2C8. Glutathione 112-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-184 8944768-7 1996 Specific-covalent binding of tienilic acid metabolites to cytochrome P-450 (incubations in the presence of 5 mM glutathione) was markedly higher upon tienilic acid oxidation by CYP 2C9 than by CYP 2C18 and CYP 2C8. Ticrynafen 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-184 8937439-3 1996 The major oxidative pathway in flurbiprofen metabolism is to a 4"-hydroxy metabolite, and recently we demonstrated that cytochrome P450 2C9 and its R144C variant were involved in this process (Tracy et al., Biochem Pharmacol 49: 1269-1275, 1995). Flurbiprofen 31-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-139 8937439-5 1996 In evaluating flurbiprofen as a potential probe for cytochrome P450 2C9, it is important to assess the involvement of additional P450s in this process. Flurbiprofen 14-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 8946475-1 1996 Cytochrome P450 CYP2C9 metabolizes a wide variety of clinically important drugs, including phenytoin, tolbutamide, warfarin and a large number of non-steroidal anti-inflammatory drugs. Phenytoin 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 8946475-1 1996 Cytochrome P450 CYP2C9 metabolizes a wide variety of clinically important drugs, including phenytoin, tolbutamide, warfarin and a large number of non-steroidal anti-inflammatory drugs. Tolbutamide 102-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 8946475-1 1996 Cytochrome P450 CYP2C9 metabolizes a wide variety of clinically important drugs, including phenytoin, tolbutamide, warfarin and a large number of non-steroidal anti-inflammatory drugs. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 8823314-2 1996 Patients with tienilic acid hepatitis exhibit autoantibodies that recognize unalkylated cytochrome P450 2C9 in humans but recognize 2C11 in rats. Ticrynafen 14-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-107 9131488-10 1996 Differences in rates of production of hydroxylamine metabolites of the drugs by cytochrome P450 (CYP2C9), myeloperoxidase, and thyroid, roxidase, along with an inherited abnormality in detoxification of the hydroxylamines are critically important in determining individual differences in adverse reaction risk. Hydroxylamine 38-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9131488-10 1996 Differences in rates of production of hydroxylamine metabolites of the drugs by cytochrome P450 (CYP2C9), myeloperoxidase, and thyroid, roxidase, along with an inherited abnormality in detoxification of the hydroxylamines are critically important in determining individual differences in adverse reaction risk. Hydroxylamines 207-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 8809086-1 1996 The purpose of the present studies was to define the role of the I359L allelic variant of CYP2C9 in the metabolism of the low therapeutic index anticoagulant warfarin, by performing in vitro kinetic studies with the two enantiomers of the drug. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 8886601-8 1996 Furthermore, rates of norfentanyl production by 10 individual human liver samples were highly correlated (r2 = 0.876, F = 56.46 p < 0.001) with immunochemically determined levels of CYP3A4 present in the samples but not with levels of CYP2C8, CYP2C9, CYP2C19, or CYP2E1. norfentanyl 22-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 246-252 8873220-0 1996 The role of the CYP2C9-Leu359 allelic variant in the tolbutamide polymorphism. Tolbutamide 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 8687505-8 1996 The precocious expression of CYP2C in SIDS could result in a higher production of epoxyeicosatrienoic acids in the neonate, believed to act as relaxant of pulmonary smooth muscles. epoxyeicosatrienoic acids 82-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-34 8700140-2 1996 Cytochrome P4502C9(CYP2C9), involved in the metabolism of tienilic acid, was shown to be a target for tienilic acid-reactive metabolites and for autoantibodies. Ticrynafen 58-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 8700140-2 1996 Cytochrome P4502C9(CYP2C9), involved in the metabolism of tienilic acid, was shown to be a target for tienilic acid-reactive metabolites and for autoantibodies. Ticrynafen 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 8886617-1 1996 The effect of branch pathways on the observed intramolecular isotope effect and deuterium retention associated with 6- and 7-hydroxylation of selectively monodeuterated (R)- and (S)-warfarin with cytochrome P450 (CYP) 2C9 and CYP1A2 were studied. Deuterium 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-221 8886617-1 1996 The effect of branch pathways on the observed intramolecular isotope effect and deuterium retention associated with 6- and 7-hydroxylation of selectively monodeuterated (R)- and (S)-warfarin with cytochrome P450 (CYP) 2C9 and CYP1A2 were studied. (r)- and (s)-warfarin 169-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-221 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. Sulfur 63-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. 1,5-anhydro-2-deoxy-d-arabino-hexitol 67-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. (s)-6d1-warfarin 76-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. (r)-7d1 156-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. (r)-6d1 169-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-2 1996 cDNA-expressed CYP2C9 was incubated with enantiomerically pure (S)-7d1- and (S)-6d1-warfarin, and expressed CYP1A2 was incubated with enantiomerically pure (R)-7d1- and (R)-6d1-warfarin. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8886617-5 1996 Deuterium isotope effects of 1.17 and 1.23 accompanied formation of 7-hydroxywarfarin from (S)-7d1-warfarin by CYP2C9 and from (R)-7d1-warfarin by CYP1A2, respectively. Deuterium 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 8886617-5 1996 Deuterium isotope effects of 1.17 and 1.23 accompanied formation of 7-hydroxywarfarin from (S)-7d1-warfarin by CYP2C9 and from (R)-7d1-warfarin by CYP1A2, respectively. 7-hydroxywarfarin 68-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 8886617-5 1996 Deuterium isotope effects of 1.17 and 1.23 accompanied formation of 7-hydroxywarfarin from (S)-7d1-warfarin by CYP2C9 and from (R)-7d1-warfarin by CYP1A2, respectively. (s)-7d1 91-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 8886617-5 1996 Deuterium isotope effects of 1.17 and 1.23 accompanied formation of 7-hydroxywarfarin from (S)-7d1-warfarin by CYP2C9 and from (R)-7d1-warfarin by CYP1A2, respectively. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 8886617-5 1996 Deuterium isotope effects of 1.17 and 1.23 accompanied formation of 7-hydroxywarfarin from (S)-7d1-warfarin by CYP2C9 and from (R)-7d1-warfarin by CYP1A2, respectively. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 8874828-1 1996 We examined the effect of CYP2C9/19 polymorphisms on the pharmacokinetics of phenytoin in 17 Japanese patients with epilepsy. Phenytoin 77-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 8873220-11 1996 Studies in a recombinant yeast expression system showed that the Leu359 variant had the highest Km and the lowest Vmac for hydroxylation of tolbutamide of all the CYP2C9 allelic variants. vmac 114-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 8873220-11 1996 Studies in a recombinant yeast expression system showed that the Leu359 variant had the highest Km and the lowest Vmac for hydroxylation of tolbutamide of all the CYP2C9 allelic variants. Tolbutamide 140-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 8873220-13 1996 The present data suggest that the incidence of the Leu359 allelic variant of CYP2C9 may account for the occurrence of poor metabolizers of tolbutamide. Tolbutamide 139-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 8681486-10 1996 LKM-2 antibodies (anti-CYP2C) caused a 75% to 100% inhibition of norantipyrine and a 58% to 80% inhibition of 3-hydroxymethylantipyrine formation. Edaravone 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-28 8681455-3 1996 In particular, the CYP102-derived structures of CYP3A4, CYP2D6 and CYP2C9 are able to rationalize the routes of tamoxifen metabolism reported in human subjects. Tamoxifen 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 8799527-11 1996 Sulphaphenazole (SPA), a CYP2C9 inhibitor, decreased the rate of NT formation in a concentration dependent manner, whereas a polyclonal rat liver CYP2C11 antibody, inhibitory for S-mephenytoin 4"-hydroxylation in humans, had no important effect on this reaction. Sulfaphenazole 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 8799527-11 1996 Sulphaphenazole (SPA), a CYP2C9 inhibitor, decreased the rate of NT formation in a concentration dependent manner, whereas a polyclonal rat liver CYP2C11 antibody, inhibitory for S-mephenytoin 4"-hydroxylation in humans, had no important effect on this reaction. Sulfaphenazole 17-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 8681486-10 1996 LKM-2 antibodies (anti-CYP2C) caused a 75% to 100% inhibition of norantipyrine and a 58% to 80% inhibition of 3-hydroxymethylantipyrine formation. 3-hydroxymethylantipyrine 110-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-28 8681486-17 1996 The CYP2C subfamily contains the predominant enzymes for norantipyrine formation, and CYP1A2 is also involved. Edaravone 57-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-9 8681486-18 1996 Formation of 3-hydroxymethylantipyrine is mediated by CYP1A2 and CYP2C9. 3-hydroxymethylantipyrine 13-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 8905247-8 1996 We propose that the following vicious circle may be involved in this interaction: induction by phenytoin of hepatic enzymes involved in the metabolism of T4 and T3, decreased serum free T4 levels causing decreased activity of hepatic NADPH cytochrome P-450 reductase, a resultant decrease in hepatic P-450 IIC9 catalyzed hydroxylation of phenytoin, increased serum phenytoin concentrations and further induction of T4 and T3 hepatic metabolism. Phenytoin 95-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-310 8793607-3 1996 Diclofenac is widely used in the treatment of rheumatic diseases and is mainly metabolized in the liver by CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 8627562-10 1996 The cDNA expressed enzymes CYP2C8, CYP2C9 and CYP2C19 catalyzed varying rates of lansoprazole 5-hydroxylation at a substrate concentration of 50 microM, but only CYPC19 catalyzed this reaction at 1 microM. Lansoprazole 81-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 8866821-1 1996 A preliminary report implicated cytochrome P450 (CYP) 2C9 in the human liver microsomal O-demethylation of S-naproxen, suggesting that this pathway may be suitable for investigation of human hepatic CYP2C9 in vitro. Naproxen 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-57 8866821-1 1996 A preliminary report implicated cytochrome P450 (CYP) 2C9 in the human liver microsomal O-demethylation of S-naproxen, suggesting that this pathway may be suitable for investigation of human hepatic CYP2C9 in vitro. Naproxen 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 199-205 8866821-2 1996 Kinetic and inhibitor studies with human liver microsomes and confirmatory investigations with cDNA-expressed enzymes were undertaken here to define the role of CYP2C9 and other isoforms in the O-demethylation of R- and S-naproxen. r- and s-naproxen 213-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 8866821-7 1996 cDNA-expressed CYP2C9 and CYP1A2 were both shown to O-demethylate R- and S-naproxen. o-demethylate r- and s-naproxen 52-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 8866821-9 1996 The data demonstrate that CYP2C9 and CYP1A2 together account for the majority of human liver R- and S-naproxen O-demethylation, precluding the use of either R- or S-naproxen as a CYP isoform-specific substrate in vitro and in vivo. r- and s-naproxen 93-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 8801060-13 1996 In contrast, amiodarone (IC50, 25 microM) and flecainide (49 microM) inhibited CYP2C9 and quinidine (0.26 microM), and flecainide (0.44 microM) inhibited CYP2D6. Amiodarone 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 8801060-13 1996 In contrast, amiodarone (IC50, 25 microM) and flecainide (49 microM) inhibited CYP2C9 and quinidine (0.26 microM), and flecainide (0.44 microM) inhibited CYP2D6. Flecainide 46-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 8861733-3 1996 Diclofenac is widely used in the treatment of rheumatic diseases and is mainly metabolized in the liver by CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 8866916-0 1996 In vitro interaction of the antipsychotic agent olanzapine with human cytochromes P450 CYP2C9, CYP2C19, CYP2D6 and CYP3A. Olanzapine 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 8866916-2 1996 The ability of olanzapine to inhibit the metabolism of marker catalytic activities for the cytochromes P450 CYP3A, CYP2D6, CYP2C9, and CYP2C19 was examined. Olanzapine 15-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 8866916-7 1996 Tolbutamide metabolism to 4-hydroxy tolbutamide, form selective for CYP2C9, was competitively inhibited by clozapine and phenytoin (Ki of 31 microM and 17 microM, respectively). Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 8866916-7 1996 Tolbutamide metabolism to 4-hydroxy tolbutamide, form selective for CYP2C9, was competitively inhibited by clozapine and phenytoin (Ki of 31 microM and 17 microM, respectively). 4'-hydroxytolbutamide 26-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 8866916-7 1996 Tolbutamide metabolism to 4-hydroxy tolbutamide, form selective for CYP2C9, was competitively inhibited by clozapine and phenytoin (Ki of 31 microM and 17 microM, respectively). Clozapine 107-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 8866916-7 1996 Tolbutamide metabolism to 4-hydroxy tolbutamide, form selective for CYP2C9, was competitively inhibited by clozapine and phenytoin (Ki of 31 microM and 17 microM, respectively). Phenytoin 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 8866916-12 1996 The calculated percent inhibition by olanzapine of substrates metabolized by CYP3A, CYP2D6, CYP2C9, and CYP2C19 was modeled assuming a total plasma concentration in the therapeutic range (0.2 microM). Olanzapine 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 8742240-4 1996 There are now a number of substrates reported that have routes of metabolism ascribed specifically to cytochrome P4502C9 (CYP2C9), the isoform mainly responsible for tolbutamide hydroxylation. Tolbutamide 166-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-120 8742240-4 1996 There are now a number of substrates reported that have routes of metabolism ascribed specifically to cytochrome P4502C9 (CYP2C9), the isoform mainly responsible for tolbutamide hydroxylation. Tolbutamide 166-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 8742240-6 1996 The substrate specificity has been rationalized in terms of a hydrogen bond donor/acceptor model and, by use of molecular modeling, an active site template model for CYP2C9 has been generated. Hydrogen 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 8542584-8 1996 We have demonstrated that the mutation frequency of ochratoxin A was increased dependent upon concentration in NIH/3T3 cell lines, stably expressing human CYP1A1, CYP1A2, CYP2C10, and CYP3A4. ochratoxin A 52-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-178 8825200-12 1996 It is concluded that the O-demethylation of naproxen (< or = 0.4 mM) is catalyzed by CYP2C subfamily members (CYP2C9/10) and CYP1A2 in human liver microsomes. Naproxen 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-93 8825200-12 1996 It is concluded that the O-demethylation of naproxen (< or = 0.4 mM) is catalyzed by CYP2C subfamily members (CYP2C9/10) and CYP1A2 in human liver microsomes. Naproxen 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-122 8613951-6 1996 Ritonavir was also found to be an inhibitor of the reactions mediated by CYP2D6 (IC50 = 2.5 microM) and CYP2C9/10 (IC50 = 8.0 microM). Ritonavir 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 8613951-7 1996 The results of this study indicate the potential for in vivo inhibition of the metabolism by ritonavir of drugs that are CYP3A, CYP2D6 and, to a lesser extent, CYP2C9/10 substrates. Ritonavir 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 8822040-2 1996 Recently, it has been shown that phenytoin and tolbutamide are metabolised by CYP2C9/10 whereas mephenytoin is metabolised by CYP2C19. Phenytoin 33-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 8822040-2 1996 Recently, it has been shown that phenytoin and tolbutamide are metabolised by CYP2C9/10 whereas mephenytoin is metabolised by CYP2C19. Tolbutamide 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 8737761-2 1996 METHODS: Affinity was determined as the inhibitory potency for prototype reactions for 3 major drug metabolising enzymes: diclofenac 4"-hydroxylation (CYP2C9), dextromethorphan O-demethylation (CYP2D6), and midazolam 1"-hydroxylation (CYP3A4). Diclofenac 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 8857077-4 1996 The apparent affinity of lornoxicam was high for CYP2C9, but negligible for CYP3A4 and CYP2D6. lornoxicam 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 8737761-4 1996 Racemic and (+)- and (-)-fluvastatin showed moderate affinity (estimated Ki > 50 micromol.1(-1)) for CYP2D6 and CYP3A4, whereas their affinity for CYP2C9 was high (estimated Ki < 1 micromol.1(-1)). (+)- and (-)-fluvastatin 12-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 8737761-6 1996 CONCLUSION: Fluvastatin selectively inhibits a major drug metabolising enzyme (CYP2C9), the (+)-isomer (pharmacologically more active) showing 4-5 fold higher affinity. Fluvastatin 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 8737761-7 1996 As already reported for lovastatin and simvastatin, in vivo drug interactions by inhibition of liver oxidation of CYP2C9 substrates (e.g. hypoglyceamic sulphonylureas and oral anticoagulants) may be expected. Lovastatin 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 8737761-7 1996 As already reported for lovastatin and simvastatin, in vivo drug interactions by inhibition of liver oxidation of CYP2C9 substrates (e.g. hypoglyceamic sulphonylureas and oral anticoagulants) may be expected. Simvastatin 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 8857077-0 1996 Role of human liver microsomal CYP2C9 in the biotransformation of lornoxicam. lornoxicam 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 8857077-2 1996 The reaction kinetics were characterised, the affinity of lornoxicam for three major human drug metabolising cytochrome P-450 isozymes (CYP2C9, CYP2D6 and CYP3A4) was determined, and inhibition of the reaction by known substrates (diclofenac, ibuprofen, mefenamic acid, phenytoin, tolbutamide and warfarin) and the prototype inhibitor (sulphaphenazole) of CYP2C9 was investigated. lornoxicam 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 8857077-2 1996 The reaction kinetics were characterised, the affinity of lornoxicam for three major human drug metabolising cytochrome P-450 isozymes (CYP2C9, CYP2D6 and CYP3A4) was determined, and inhibition of the reaction by known substrates (diclofenac, ibuprofen, mefenamic acid, phenytoin, tolbutamide and warfarin) and the prototype inhibitor (sulphaphenazole) of CYP2C9 was investigated. lornoxicam 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 356-362 8857077-5 1996 Inhibition of lornoxicam 5"-hydroxylation by CYP2C9 substrates and sulphaphenazole competitively and completely inhibited lornoxicam 5"-hydroxylation (Ki = 0.31 mu mol center dot l-1 as well as lornoxicam clearance (Ki = 0.33 mu mol center dot l-1), partial metabolic clearance (fm) = 0.95). lornoxicam 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 8857077-5 1996 Inhibition of lornoxicam 5"-hydroxylation by CYP2C9 substrates and sulphaphenazole competitively and completely inhibited lornoxicam 5"-hydroxylation (Ki = 0.31 mu mol center dot l-1 as well as lornoxicam clearance (Ki = 0.33 mu mol center dot l-1), partial metabolic clearance (fm) = 0.95). lornoxicam 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 8857077-5 1996 Inhibition of lornoxicam 5"-hydroxylation by CYP2C9 substrates and sulphaphenazole competitively and completely inhibited lornoxicam 5"-hydroxylation (Ki = 0.31 mu mol center dot l-1 as well as lornoxicam clearance (Ki = 0.33 mu mol center dot l-1), partial metabolic clearance (fm) = 0.95). lornoxicam 122-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 8857077-6 1996 CONCLUSION: 5"-Hydroxylation appears to be the only cytochrome P-450 catalysed metabolic reaction of lornoxicam by human liver microsomes and this major in vivo biotransformation pathway is catalysed virtually exclusively by CYP2C9. lornoxicam 101-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 225-231 8750925-8 1995 Anti-LKM2 (anti-liver/kidney microsome) autoantibodies (which inhibit CYP2C9 and 2C19) and sulfaphenazole (a specific CYP2C9 inhibitor) reduced the maximum rate of formation of D-703 by 81.5 +/- 4.5% and 45%, that of D-702 by 52.7 +/- 7.5% and 72.5%, respectively. D 703 177-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 8698676-1 1996 The article focuses on the effects of the serotonin selective reuptake inhibitors (SSRIs) on specific drug metabolizing isoenzymes: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. Serotonin 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 8703658-9 1995 Studies with low molecular weight inhibitors illustrate the importance of CYP2C9 and CYP3A in dapsone N-hydroxylation. Dapsone 94-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 8750925-8 1995 Anti-LKM2 (anti-liver/kidney microsome) autoantibodies (which inhibit CYP2C9 and 2C19) and sulfaphenazole (a specific CYP2C9 inhibitor) reduced the maximum rate of formation of D-703 by 81.5 +/- 4.5% and 45%, that of D-702 by 52.7 +/- 7.5% and 72.5%, respectively. Sulfaphenazole 91-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 8747411-0 1995 Genetic polymorphism of CYP2C9 and its effect on warfarin maintenance dose requirement in patients undergoing anticoagulation therapy. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 8654205-5 1995 The turnover rates (min-1) of DETC-Me sulfoxidation by the cDNA-expressed cytochrome P450 enzymes ranked as follows: CYP3A4 > CYP2A6 = CYP2C9 > CYP1A2 > CYP2B6 = CYP2E1 > CYP1A1 > CYP2D6. detc-me 30-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 7586933-0 1995 In vivo inhibition profile of cytochrome P450TB (CYP2C9) by (+/-)-fluvastatin. Fluvastatin 60-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 7586933-1 1995 BACKGROUND: (+/-)-Fluvastatin is a synthetic 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor that selectively and competitively inhibits P450TB (CYP2C9) in vitro. Fluvastatin 12-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 8587855-3 1995 This review summarizes information to date regarding the inhibitory potency of fluoxetine, fluvoxamine, paroxetine, sertraline, nefazodone, and venlafaxine on five isoenzyme systems: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. Fluoxetine 79-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 8587855-3 1995 This review summarizes information to date regarding the inhibitory potency of fluoxetine, fluvoxamine, paroxetine, sertraline, nefazodone, and venlafaxine on five isoenzyme systems: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. Sertraline 116-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 8587855-3 1995 This review summarizes information to date regarding the inhibitory potency of fluoxetine, fluvoxamine, paroxetine, sertraline, nefazodone, and venlafaxine on five isoenzyme systems: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. nefazodone 128-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 8587855-3 1995 This review summarizes information to date regarding the inhibitory potency of fluoxetine, fluvoxamine, paroxetine, sertraline, nefazodone, and venlafaxine on five isoenzyme systems: CYP2D6, CYP3A3/4, CYP1A2, CYP2C9, and CYP2C19. Venlafaxine Hydrochloride 144-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 8654202-5 1995 In addition, the respective omega-OH finasteride and omega-oic acid finasteride metabolites were generated only by microsomes containing recombinant CYP3A4, but not the other isozymes (CYP1A1, CYP2B6, CYP2C8, CYP2C9, CYP2D6, and CYP2E1). omega-oh finasteride 28-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 7574697-10 1995 Compared to Cyp 2C8, arachidonic acid epoxidation by Cyp 2C10 was less regio- and stereo-selective and generated mixtures of 8(S), 9(R)-, 11(S), 12(R)-, and 14(R), 15(S)-epoxyeicosatrienoic acids (with optical purities of 66, 69, 63%, respectively). Arachidonic Acid 21-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-61 8654202-5 1995 In addition, the respective omega-OH finasteride and omega-oic acid finasteride metabolites were generated only by microsomes containing recombinant CYP3A4, but not the other isozymes (CYP1A1, CYP2B6, CYP2C8, CYP2C9, CYP2D6, and CYP2E1). omega-oic acid finasteride 53-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 8654206-3 1995 Results suggested that whereas the metabolism of zileuton and Abbott-66193 were mediated by the same CYP forms, the CYP forms responsible for hydroxylation (CYP1A2 and CYP2C9/10) were distinct from those involved in sulfoxidation (CYP3A > CYP2C9/10). zileuton 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 242-248 8654206-6 1995 On the other hand, hydroxylation of the two compounds (zileuton, Km = 0.34 +/- 0.25 mM, Vmax = 17.8 +/- 5.58 pmol/min/mg; Abbott-66193,Km = 0.39 +/- 0.14 mM, Vmax = 1061 +/- 220 pmol/min/mg) was significantly correlated with 7-ethoxyresorufin O-deethylase (CYP1A2; r=0.652-0.762; p<0.01, N=11) and tolbutamide methyl hydroxylase (CYP2C9/10; r=0.863-0.935; p<0.01, N=10) activity in human liver microsome, and was inhibited (26-51%) by well-known CYP1A2 inhibitors (furafylline and alpha-naphthoflavone). zileuton 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 333-339 8654206-8 1995 Abbott-66193 was a better substrate for CYP2C9/10, when compared with zileuton: 1) the effect of sulfaphenazole on hydroxylation in human liver microsomes was more pronounced for Abbott-66193 than zileuton (56% vs. 9% inhibition); and 2) the rate of Abbott-66193 hydroxylation by purified CYP2C9 was almost 30-fold greater than that of zilueton. Sulfaphenazole 97-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 7574697-10 1995 Compared to Cyp 2C8, arachidonic acid epoxidation by Cyp 2C10 was less regio- and stereo-selective and generated mixtures of 8(S), 9(R)-, 11(S), 12(R)-, and 14(R), 15(S)-epoxyeicosatrienoic acids (with optical purities of 66, 69, 63%, respectively). Sulfur 126-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-61 7574697-10 1995 Compared to Cyp 2C8, arachidonic acid epoxidation by Cyp 2C10 was less regio- and stereo-selective and generated mixtures of 8(S), 9(R)-, 11(S), 12(R)-, and 14(R), 15(S)-epoxyeicosatrienoic acids (with optical purities of 66, 69, 63%, respectively). (r) 132-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-61 7574697-10 1995 Compared to Cyp 2C8, arachidonic acid epoxidation by Cyp 2C10 was less regio- and stereo-selective and generated mixtures of 8(S), 9(R)-, 11(S), 12(R)-, and 14(R), 15(S)-epoxyeicosatrienoic acids (with optical purities of 66, 69, 63%, respectively). (s)-epoxyeicosatrienoic acids 166-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-61 7614688-7 1995 Treatment of cynomolgus monkeys with rifampicin induced hepatic cytochromes P450 related to human CYP3A4 and CYP2C9/10 without inducing CYP1A1 or CYP1A2. Rifampin 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 7654690-0 1995 The substrate binding site of human liver cytochrome P450 2C9: an approach using designed tienilic acid derivatives and molecular modeling. Ticrynafen 90-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-61 7654690-2 1995 The ability of 10 2-aroylthiophenes related to tienilic acid to act as substrates for CYP2C9 was studied. 2-aroylthiophenes 18-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 7654690-2 1995 The ability of 10 2-aroylthiophenes related to tienilic acid to act as substrates for CYP2C9 was studied. Ticrynafen 47-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Carboxylic Acids 70-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Carboxylic Acids 70-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Phenol 87-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). 1H-tetrazole 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). 1H-tetrazole 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Ticrynafen 149-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Suprofen 195-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Suprofen 195-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 7654690-8 1995 In fact, the four new substrates of CYP2C9 described in this study, a carboxylic acid, phenol, and tetrazole derivative, each of which is related to tienilic acid, and the antiinflammatory drug, suprofen (with Km between 12 and 130 microM and kcat between 0.2 and 1.3 min-1), as well as almost all CYP2C9 substrates reported in the literature, exhibit a pKa below 7 (except phenytoin whose pKa is 8.1). Phenytoin 374-383 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 7654690-10 1995 By using molecular modeling techniques, 12 CYP2C9 substrates were superimposed with respect to their hydroxylation site and fitted onto templates, which were rigid molecules such as (S)-warfarin and phenytoin. Warfarin 182-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 7654690-10 1995 By using molecular modeling techniques, 12 CYP2C9 substrates were superimposed with respect to their hydroxylation site and fitted onto templates, which were rigid molecules such as (S)-warfarin and phenytoin. Phenytoin 199-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 8535411-0 1995 Involvement of CYP2C in the metabolism of cannabinoids by human hepatic microsomes from an old woman. Cannabinoids 42-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-20 7614688-12 1995 Since cytochromes P4503A and/or P4502C are constitutively expressed in cynomolgus monkey hepatic microsomes, and upon induction with rifampicin are associated with an increased metabolic activation of IQ but not MeIQx, it appears that CYP3A and/or CYP2C are the isoform(s) showing the selective substrate specificity in the metabolic activation of IQ over MeIQx. Rifampin 133-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 248-253 7625847-0 1995 Arachidonic acid metabolism by human cytochrome P450s 2C8, 2C9, 2E1, and 1A2: regioselective oxygenation and evidence for a role for CYP2C enzymes in arachidonic acid epoxygenation in human liver microsomes. Arachidonic Acid 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-138 7625847-0 1995 Arachidonic acid metabolism by human cytochrome P450s 2C8, 2C9, 2E1, and 1A2: regioselective oxygenation and evidence for a role for CYP2C enzymes in arachidonic acid epoxygenation in human liver microsomes. Arachidonic Acid 150-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-138 7625847-17 1995 The CYP2C inhibitor sulfaphenazole depressed human liver microsomal epoxygenase activity by 50% at 50 microM, while alpha-naphthoflavone inhibited arachidonic acid epoxygenase activity by 27% at 2 microM and by 32% at 10 microM. Sulfaphenazole 20-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-9 7625847-18 1995 Collectively, these findings suggest that human liver microsomal arachidonic acid metabolism is catalyzed principally by CYP2C enzymes. Arachidonic Acid 65-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-126 7614688-16 1995 Furthermore, the poor metabolic activation of MeIQx by CYP3A and CYP2C, coupled with low constitutive levels of CYP1A isozymes, provide a metabolic explanation for the low hepatocarcinogenic potency of MeIQx in cynomolgus monkeys. 2-amino-3,8-dimethylimidazo(4,5-f)quinoxaline 46-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-70 7614688-16 1995 Furthermore, the poor metabolic activation of MeIQx by CYP3A and CYP2C, coupled with low constitutive levels of CYP1A isozymes, provide a metabolic explanation for the low hepatocarcinogenic potency of MeIQx in cynomolgus monkeys. 2-amino-3,8-dimethylimidazo(4,5-f)quinoxaline 202-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-70 7794915-6 1995 Gel shift analysis demonstrated that nuclear proteins from HepG2 cells had a high binding affinity for a 20-bp oligonucleotide containing the HPF-1 site of CYP2C9. Oligonucleotides 111-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 7763308-0 1995 Role of cytochrome P450 2C9 and an allelic variant in the 4"-hydroxylation of (R)- and (S)-flurbiprofen. (r)- and (s)-flurbiprofen 78-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-27 7741772-1 1995 58C80 [2-(4-t-butylcyclohexyl)-3-hydroxy-1,4-naphthoquinone] is an experimental naphthoquinone antimalarial drug which undergoes extensive alkyl hydroxylation to a single t-butylhydroxy metabolite in man in vivo and also in human liver microsomes, where this is catalysed primarily by a 54 kDa CYP2C9 form of cytochrome P450, P450hB20-27. Bupravaquone 7-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 294-300 7741772-1 1995 58C80 [2-(4-t-butylcyclohexyl)-3-hydroxy-1,4-naphthoquinone] is an experimental naphthoquinone antimalarial drug which undergoes extensive alkyl hydroxylation to a single t-butylhydroxy metabolite in man in vivo and also in human liver microsomes, where this is catalysed primarily by a 54 kDa CYP2C9 form of cytochrome P450, P450hB20-27. Naphthoquinones 45-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 294-300 7736913-8 1995 Inhibitors of CYP3A4/5 (gestodene and ketoconazole) and CYP2C9/10 (sulfaphenazole) attenuated the oxidation of both substrates. Sulfaphenazole 67-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 7891318-0 1995 Human hepatic cytochrome P450 2C9 catalyzes the rate-limiting pathway of torsemide metabolism. Torsemide 73-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-33 7891318-5 1995 The microsomal reaction was almost completely abolished by the specific CYP2C9 inhibitor sulfaphenazole and was inhibited competitively by the alternative CYP2C9 substrate tolbutamide. Sulfaphenazole 89-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 7891318-5 1995 The microsomal reaction was almost completely abolished by the specific CYP2C9 inhibitor sulfaphenazole and was inhibited competitively by the alternative CYP2C9 substrate tolbutamide. Sulfaphenazole 89-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 7891318-5 1995 The microsomal reaction was almost completely abolished by the specific CYP2C9 inhibitor sulfaphenazole and was inhibited competitively by the alternative CYP2C9 substrate tolbutamide. Tolbutamide 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 7891318-5 1995 The microsomal reaction was almost completely abolished by the specific CYP2C9 inhibitor sulfaphenazole and was inhibited competitively by the alternative CYP2C9 substrate tolbutamide. Tolbutamide 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 7891318-6 1995 Torsemide tolyl methylhydroxylase activity in microsomes from 16 human livers correlated significantly (rs = .81-.88) with tolbutamide and phenytoin hydroxylation, both CYP2C9-mediated reactions. Torsemide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 7891318-7 1995 Complementary DNA-expressed CYP2C9 catalyzed torsemide tolyl methylhydroxylation with an apparent Km (23 microM) similar to that observed for human liver microsomes and the IC50 values for sulfaphenazole inhibition of the reaction were essentially identical for the two enzyme sources. Torsemide 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 7891318-7 1995 Complementary DNA-expressed CYP2C9 catalyzed torsemide tolyl methylhydroxylation with an apparent Km (23 microM) similar to that observed for human liver microsomes and the IC50 values for sulfaphenazole inhibition of the reaction were essentially identical for the two enzyme sources. Sulfaphenazole 189-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 7891318-8 1995 Taken together, these data demonstrate that human hepatic torsemide tolyl methylhydroxylation is catalyzed predominantly, if not solely, by CYP2C9. Torsemide 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 7628308-7 1995 In a panel of human microsomes, SMX-HA formation correlated with tolbutamide hydroxylase activity (r = 0.75; p = 0.01); CYP2C9 content (r = 0.79; p < 0.01) and was inhibited 70% by 500 microM tolbutamide and 90% by 100 microM sulfaphenazole. sulfamethoxazole hydroxylamine 32-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 7628308-8 1995 Recombinant CYP2C9 catalyzed the N-hydroxylation of SMX. Sulfamethoxazole 52-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 7628308-9 1995 SMX-HA formation in human hepatic microsomes was therefore mediated predominantly by CYP2C9. Sulfamethoxazole 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 7736913-9 1995 It was then demonstrated that microsomes containing either recombinant human liver CYP2C9 or CYP3A4 were capable of oxidizing both losartan and the aldehyde E3179 to the carboxylic acid E3174. Losartan 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 7736913-9 1995 It was then demonstrated that microsomes containing either recombinant human liver CYP2C9 or CYP3A4 were capable of oxidizing both losartan and the aldehyde E3179 to the carboxylic acid E3174. Aldehydes 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 7736913-9 1995 It was then demonstrated that microsomes containing either recombinant human liver CYP2C9 or CYP3A4 were capable of oxidizing both losartan and the aldehyde E3179 to the carboxylic acid E3174. Carboxylic Acids 170-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 7736913-10 1995 Subsequently, it was shown that rabbit anti-CYP2C9 and anti-CYP3A3/4 inhibited the oxidation of losartan to E3174 in incubations with human liver microsomes. Losartan 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 7736913-10 1995 Subsequently, it was shown that rabbit anti-CYP2C9 and anti-CYP3A3/4 inhibited the oxidation of losartan to E3174 in incubations with human liver microsomes. losartan carboxylic acid 108-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 7720520-5 1995 The probes sulfaphenazole (CYP2C9) and quinidine (CYP2D6) selectively inhibited tolbutamide methylhydroxylation and bufuralol 1"-hydroxylation, respectively. Sulfaphenazole 11-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 7773301-4 1995 The T416-->C mutation in exon 3 of CYP2C9 (Cys144-->Arg) creates an Ava II site. Arginine 58-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 7773301-13 1995 The G1236-->A mutation in exon 8 of CYP2C9 (Gly417-->Asp) creates a Hph I site. Aspartic Acid 59-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 7720520-5 1995 The probes sulfaphenazole (CYP2C9) and quinidine (CYP2D6) selectively inhibited tolbutamide methylhydroxylation and bufuralol 1"-hydroxylation, respectively. Tolbutamide 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 7720520-5 1995 The probes sulfaphenazole (CYP2C9) and quinidine (CYP2D6) selectively inhibited tolbutamide methylhydroxylation and bufuralol 1"-hydroxylation, respectively. bufuralol 116-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 8203907-8 1994 Overall, these results establish that (a) TAO is a selective inhibitor of the human CYP3A subfamily; (b) ANF potently inhibits CYP2C8 and CYP2C9, in addition to CYPs 1A1 and 1A2; and (c) DDC cannot be employed as a diagnostic inhibitory probe for CYP2E1. Troleandomycin 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 7704034-4 1994 CYP2C9 is important in the metabolism certain of therapeutically used drugs including the anticoagulant drug warfarin and a number of nonsteroidal antiinflammatory drugs. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 7704038-5 1994 An antibody raised against CYP2C9 (anti-human CYP2C) strongly inhibited diazepam N-demethylation in EM liver microsomes at a low substrate concentration (20 microM). Diazepam 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 7704038-5 1994 An antibody raised against CYP2C9 (anti-human CYP2C) strongly inhibited diazepam N-demethylation in EM liver microsomes at a low substrate concentration (20 microM). Diazepam 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-32 7981013-11 1994 Studies with a series of CYP isoform selective inhibitors and with an inhibitory anti-CYP2C antibody indicated that temazepam formation was carried out mainly by CYP3A isoforms, whereas the formation of N-desmethyldiazepam was mediated by both CYP3A isoforms and S-mephenytoin hydroxylase. Temazepam 116-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-91 7981013-11 1994 Studies with a series of CYP isoform selective inhibitors and with an inhibitory anti-CYP2C antibody indicated that temazepam formation was carried out mainly by CYP3A isoforms, whereas the formation of N-desmethyldiazepam was mediated by both CYP3A isoforms and S-mephenytoin hydroxylase. Nordazepam 203-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-91 8375128-1 1993 V. In vivo CYP2C9 activity as probed by (S)-warfarin is not enhanced in cystic fibrosis. Warfarin 40-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 7903909-8 1994 Conversely, the formation of metabolite M5 remained unaffected by antibodies against CYP3A and by CYP3A substrates but was sensitive to diazepam inhibition, a preferential substrate of CYP2C. Diazepam 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 185-190 8148870-0 1993 Cytochrome P450 mediated metabolism of diazepam in human and rat: involvement of human CYP2C in N-demethylation in the substrate concentration-dependent manner. Diazepam 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-92 8148870-0 1993 Cytochrome P450 mediated metabolism of diazepam in human and rat: involvement of human CYP2C in N-demethylation in the substrate concentration-dependent manner. Nitrogen 96-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-92 8148870-5 1993 Inhibitions of the N-demethylation by anti-CYP2C antibody and S-mephenytoin also depended on the substrate concentration and was detectable only at a low substrate concentration. Nitrogen 19-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-48 8216369-0 1993 Cytochrome P450 2C9 is responsible for hydroxylation of the naphthoquinone antimalarial drug 58C80 in human liver. Naphthoquinones 60-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 8004131-0 1994 Impaired (S)-warfarin metabolism catalysed by the R144C allelic variant of CYP2C9. Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 8286335-0 1994 Thiophene derivatives as new mechanism-based inhibitors of cytochromes P-450: inactivation of yeast-expressed human liver cytochrome P-450 2C9 by tienilic acid. Thiophenes 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-142 8286335-0 1994 Thiophene derivatives as new mechanism-based inhibitors of cytochromes P-450: inactivation of yeast-expressed human liver cytochrome P-450 2C9 by tienilic acid. Ticrynafen 146-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-142 8333835-1 1993 There is a genetic polymorphism in humans in the metabolism of S-mephenytoin which has been suggested to be mediated by either CYP2C18 or CYP2C9. Mephenytoin 63-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 8333835-2 1993 We have isolated genomic clones for CYP2C9 and CYP2C18 from the liver of an individual phenotyped in vitro as an extensive metabolizer of S-mephenytoin. Mephenytoin 138-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 8518837-6 1993 Furthermore, CYP2C9 produced by cDNA expression in yeasts, catalysed tolbutamide hydroxylation at rates similar to the rapid metabolizer group at both the 0.1 mM and 2.4 mM concentrations. Tolbutamide 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 8333835-7 1993 The availability of the sequences of the upstream regions and intron-exon junctions of CYP2C9 and CYP2C18 will allow future analysis of these genes in humans which differ in their ability to metabolize S-mephenytoin and other drugs. Sulfur 202-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 8518837-8 1993 Clear correlations were observed between the rate of microsomal tolbutamide hydroxylation at 0.1 mM and CYP2C9 protein content or the rate of S-mephenytoin 4"-hydroxylation in human liver. Tolbutamide 64-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 8518837-9 1993 These results indicate that considerable portions of microsomal tolbutamide hydroxylation are catalysed by CYP2C9 or the closely related form in the rapid metabolizers. Tolbutamide 64-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 2200675-5 1990 Phenobarbital and rifampicin increased P-450 IIC8/9/10 mRNA transcripts and the corresponding protein, while 3-methylcholanthrene was ineffective. Phenobarbital 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-54 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Debrisoquin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Sparteine 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 8454228-2 1993 Debrisoquine/sparteine and mephenytoin polymorphisms are now known to be derived from defects in the human liver of specific forms of microsomal CYP (P450 or previously termed cytochrome P-450), CYP2D6 and CYP2C9 (2C18), respectively. Mephenytoin 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 1445376-5 1992 The expressed enzymes showed tolbutamide hydroxylase activities, and these activities were inhibited by antibodies against P-450-HM2, a probable CYP2C9. p-450-hm2 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 8070855-4 1993 Of the 10 human P450 forms studied, CYP2B6 was the most effective in forming of styrene glycol, followed by CYP1A2, CYP2E1 and CYP2C8; the human P450s CYP3A3, CYP3A4 and CYP3A5 also catalysed metabolism, but were much less active; and CYP2A6, CYP2C9 and CYP2D6 had little detectable activity. styrene glycol 80-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 243-249 8417277-0 1993 Cytochrome P450TB (CYP2C): a major monooxygenase catalyzing diclofenac 4"-hydroxylation in human liver. Diclofenac 60-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-24 8417277-2 1993 Inhibition studies were performed with tolbutamide and sulfaphenazole (respectively the prototype substrate and a selective inhibitor of cytochrome P450TB--CYP2C subfamily), and with phenytoin and (+/-)-warfarin, other proposed substrates of P450TB. Sulfaphenazole 55-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-161 8417277-2 1993 Inhibition studies were performed with tolbutamide and sulfaphenazole (respectively the prototype substrate and a selective inhibitor of cytochrome P450TB--CYP2C subfamily), and with phenytoin and (+/-)-warfarin, other proposed substrates of P450TB. Terbium 152-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-161 8417277-6 1993 Diclofenac 4"-hydroxylation is predominantly catalyzed by a cytochrome P450 isozyme of the CYP2C subfamily, most likely CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-96 8417277-6 1993 Diclofenac 4"-hydroxylation is predominantly catalyzed by a cytochrome P450 isozyme of the CYP2C subfamily, most likely CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 1917894-0 1991 Cytochrome P-450 human-2 (P-450IIC9) in mephenytoin hydroxylation polymorphism in human livers: differences in substrate and stereoselectivities among microheterogeneous P-450IIC species expressed in yeasts. Mephenytoin 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-35 2200675-5 1990 Phenobarbital and rifampicin increased P-450 IIC8/9/10 mRNA transcripts and the corresponding protein, while 3-methylcholanthrene was ineffective. Rifampin 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-54 20702214-3 1990 Phenobarbital and rifampicin were found to increase the levels of P-450 IIC8, 9, 10 mRNA and protein while troleandomycin and 3-methylcholanthrene were ineffective. Phenobarbital 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-79 20702214-3 1990 Phenobarbital and rifampicin were found to increase the levels of P-450 IIC8, 9, 10 mRNA and protein while troleandomycin and 3-methylcholanthrene were ineffective. Rifampin 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-79 33822485-2 2021 Ivosidenib is an inducer of the CYP2B6, CYP2C8, CYP2C9, and CYP3A4 and an inhibitor of P-glycoprotein (P-gp), organic anion transporting polypeptide-1B1/1B3 (OATP1B1/1B3), and organic anion transporter-3 (OAT3) in vitro. ivosidenib 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 33822485-7 2021 The model was also used to predict the DDIs of ivosidenib with CYP2B6, CYP2C8, CYP2C9, P-gp, OATP1B1/1B3, and OAT3 substrates. ivosidenib 47-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 33802239-6 2021 We found that lignans with one or two methylenedioxyphenyl groups inhibited CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP2E1 activities in a time- and concentration-dependent like their CYP3A inhibition. Lignans 14-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 33811620-0 2021 Influence of CYP2C9, VKORC1, and CYP4F2 polymorphisms on the pharmacodynamic parameters of warfarin: a cross-sectional study. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 33811620-9 2021 Groups" II and III CYP2C9 and VKORC1 genotypes were observed with reduced stable warfarin dose, increased WSI, higher log-INR variability, and increased bleeding risk. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 33811620-10 2021 The presence of *2 or *3 allele in CYP2C9 was observed with reduced stable warfarin doses akin to the presence of T alleles in VKORC1; however, the doses increased with T alleles in CYP4F2. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 33811620-12 2021 Evaluating CYP2C9, VKORC1, and CYP4F2 genetic polymorphisms prior to warfarin initiation is likely to optimize therapeutic response. Warfarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 33814360-8 2021 Voriconazole is metabolized and is a strong inhibitor of cytochrome (CYP)3A4, CYP2C9/10 and CYP2C19. Voriconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 33802239-10 2021 This suggests that the time-dependent inhibition of gomisin A against CYP2C8, CYP2C9, and CYP3A4 is due to the production of carbene reactive metabolite. schizandrol B 52-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 33802239-10 2021 This suggests that the time-dependent inhibition of gomisin A against CYP2C8, CYP2C9, and CYP3A4 is due to the production of carbene reactive metabolite. carbene 125-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 33588415-3 2020 We investigated the effect of sEH inhibitor (TPPU) on the expression of villin, CYP2C8, CYP2C9, CYP2J2, and sEH in undifferentiated and in vitro differentiated HT-29 and Caco2 cell lines. TPPU 45-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 33780197-2 2020 The most important factor predicting daily warfarin dose is individual genotype, polymorphisms of genes CYP2C9 (warfarin metabolism) and VKORC1 (sensitivity for warfarin). Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 33780197-2 2020 The most important factor predicting daily warfarin dose is individual genotype, polymorphisms of genes CYP2C9 (warfarin metabolism) and VKORC1 (sensitivity for warfarin). Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 33780197-2 2020 The most important factor predicting daily warfarin dose is individual genotype, polymorphisms of genes CYP2C9 (warfarin metabolism) and VKORC1 (sensitivity for warfarin). Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 25385663-1 2015 Genotypes for cytochrome P450 (CYP) 2C9 and vitamin K epoxide reductase complex 1 (VKORC1) contribute significantly to the inter-patient variability in warfarin dose requirements. Warfarin 152-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-39 28371488-10 2018 This study is vital to clinical usage of warfarin not only because of the pharmacokinetic parameters of the 2 pharmaceutical dosage forms of warfarin but also to obtain data on the prevalence of CYP2C9 and VKORC1 genes and their influence on the concentration of warfarin. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 29368402-3 2018 The parameters for cytochrome P450 (CYP) 3A and CYP2C9 induction by rifampicin were similarly optimized using clinical DDI data with midazolam and tolbutamide as probe substrates, respectively. Rifampin 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 9686881-0 1998 Chiral phase analysis of warfarin enantiomers in patient plasma in relation to CYP2C9 genotype. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 24002148-10 2013 Efinaconazole is a CYP inhibitor like other azole antifungals, and its lowest ki is 91 ng/mL for CYP2C9, a >130-fold higher concentration than the mean steady state Cmax observed in patients. efinaconazole 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 24002148-10 2013 Efinaconazole is a CYP inhibitor like other azole antifungals, and its lowest ki is 91 ng/mL for CYP2C9, a >130-fold higher concentration than the mean steady state Cmax observed in patients. Azoles 8-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 9686881-7 1998 Two warfarin patients homozygous for the mutant CYP2C9*2 and CYP2C9*3 alleles exhibited elevated S/R ratios relative to the mean for individuals homozygous for the wild-type CYP2C9*1 allele. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 9686881-7 1998 Two warfarin patients homozygous for the mutant CYP2C9*2 and CYP2C9*3 alleles exhibited elevated S/R ratios relative to the mean for individuals homozygous for the wild-type CYP2C9*1 allele. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 9686881-7 1998 Two warfarin patients homozygous for the mutant CYP2C9*2 and CYP2C9*3 alleles exhibited elevated S/R ratios relative to the mean for individuals homozygous for the wild-type CYP2C9*1 allele. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 9686881-8 1998 This method is suitable for population studies aimed at establishing the effect of polymorphic expression of CYP2C9 alleles on S-warfarin elimination in humans. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 34078115-6 2022 IL-6 inhibitors (sirukumab, tocilizumab, sarilumab) significantly enhance metabolism via CYP2C9 (s-warfarin), CYP2C19 (omeprazole), and CYP3A4 (simvastatin, midazolam) and reduce metabolism via CYP1A2 (caffeine). Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 17646282-4 2007 Incubating (R)-tofisopam with recombinant cytochrome P450 (P450) or with human liver microsomes and isoform-selective P450 chemical inhibitors indicated that M3 was primarily catalyzed by CYP2C9. tofisopam 11-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 34671112-5 2022 Seventy-three percent had normal functioning SLCO1B1 transporter, 4% carried the HLA-B*58:01 risk variant, and 35% carried VKORC1 and CYP2C9 variants that increased warfarin sensitivity. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 34656068-1 2022 Sulfonylureas (SUs) are predominantly metabolized by cytochrome p450 2C9 (CYP2C9) and cytochrome p450 2C19 (CYP2C19) enzymes. Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 34656068-1 2022 Sulfonylureas (SUs) are predominantly metabolized by cytochrome p450 2C9 (CYP2C9) and cytochrome p450 2C19 (CYP2C19) enzymes. Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 34656068-1 2022 Sulfonylureas (SUs) are predominantly metabolized by cytochrome p450 2C9 (CYP2C9) and cytochrome p450 2C19 (CYP2C19) enzymes. Sulfonylurea Compounds 15-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 34656068-1 2022 Sulfonylureas (SUs) are predominantly metabolized by cytochrome p450 2C9 (CYP2C9) and cytochrome p450 2C19 (CYP2C19) enzymes. Sulfonylurea Compounds 15-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 34656068-2 2022 CYP2C9 polymorphisms are associated with greater treatment response and hypoglycemic risk in SU users. Sulfonylurea Compounds 93-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34453556-1 2022 PURPOSE: The purpose of this paper is to study the correlation between demographic and clinical factors and warfarin dose of patients in Chinese Han population taking warfarin and study gene polymorphisms impact of related gene loci (CYP2C9*3, VKORC1-1639G > A) on warfarin doses, to establish a model to predict initial standard dose and maintenance dose based on CYP2C9*3, VKORC1-1639G > A genotype. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 234-240 34920277-2 2022 We investigated the role of SNPs in CYP2C9, CYP2C19, CYP2D6, UGT2B15, and UGT1A7 genes in the biotransformation of phthalates (DEHP, DEP, DiBP, DnBP, BBzP, DiNP, DidP) and DINCH by determining their urine metabolites. phthalic acid 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 34920277-6 2022 The most pronounced was the influence of CYP2C9*2 and *3 on the reduced DEHP biotransformation, with lower levels of metabolites and their ratios in men and women. Diethylhexyl Phthalate 72-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 34920277-10 2022 Our observations confirm the effect of CYP2C9*2 and *3 SNPs towards reduced DEHP biotransformation. Diethylhexyl Phthalate 76-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 34453556-1 2022 PURPOSE: The purpose of this paper is to study the correlation between demographic and clinical factors and warfarin dose of patients in Chinese Han population taking warfarin and study gene polymorphisms impact of related gene loci (CYP2C9*3, VKORC1-1639G > A) on warfarin doses, to establish a model to predict initial standard dose and maintenance dose based on CYP2C9*3, VKORC1-1639G > A genotype. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 365-371 34453556-1 2022 PURPOSE: The purpose of this paper is to study the correlation between demographic and clinical factors and warfarin dose of patients in Chinese Han population taking warfarin and study gene polymorphisms impact of related gene loci (CYP2C9*3, VKORC1-1639G > A) on warfarin doses, to establish a model to predict initial standard dose and maintenance dose based on CYP2C9*3, VKORC1-1639G > A genotype. Warfarin 265-273 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 234-240 34453556-4 2022 RESULTS: The initial dose of warfarin (mg/day) was calculated as (1.346 + 0.350 x (VKORC1-1639G > A) - 0.273 x (CYP2C9*3) + 0.245 x (body surface area) - 0.003 x (age) - 0.036 x (amine-iodine) + 0.021 x (sex))2. Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 34453556-6 2022 The maintenance dose of warfarin (mg/day) was calculated as (1.336 + 0.299 x (VKORC1-1639G > A) + 0.480 x (body surface area) - 0.214 x (CYP2C9*3) - 0.074 x (amine-iodine) - 0.003 x (age) - 0.077 x (statins) - 0.002 x (height))2. Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 34872459-11 2021 Patients with tuberculosis with older age and genetic polymorphism of CYP3A4, CYP2C9, and CYP2C19 who received long-term rifampicin treatment were more likely to have antituberculosis drug-induced liver injury. Rifampin 121-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 34780725-4 2022 We also confirmed dabrafenib as a CYP2C8, CYP2C9, CYP3A4, and CYP3A5 inhibitor. dabrafenib 18-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 34865639-3 2021 METHODS: Preclinical evaluation of strasseriolides A-D was performed by MTT-based cytotoxicity assay in THLE-2 (CRL-2706) liver cells, cardiotoxicity screening using the FluxOR potassium assay in hERG expressed HEK cells, LC-MS-based analysis of drug-drug interaction involving CYP3A4, CYP2D6 and CYP2C9 isoforms inhibition and metabolic stability assays in human liver microsomes. strasseriolides 35-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 34748821-0 2021 CYP2C9 and 3A4 play opposing roles in bioactivation and detoxification of diphenylamine NSAIDs. Diphenylamine 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34748821-8 2021 When considering hepatic CYP abundance, CYP2C9 almost exclusively accounted for diphenylamine NSAID bioactivations, whereas CYP3A4 provided a critical counterbalance favoring their overall detoxification. Diphenylamine 80-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 34896433-5 2022 In addition, alisertib was characterized as a low- to moderate-affinity inhibitor of recombinant CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2D6 isoenzymes, but without potential clinical relevance. MLN 8237 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 34952963-0 2022 Effects of CYP2C9*3 and *13 alleles on the pharmacokinetics and pharmacodynamics of glipizide in healthy Korean subjects. Glipizide 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 34952963-3 2022 In this study, we investigated the effects of CYP2C9*3 and *13 variant alleles on the pharmacokinetics and pharmacodynamics of glipizide. Glipizide 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 34952963-7 2022 The results represented that CYP2C9*3 and *13 alleles significantly affected the pharmacokinetics of glipizide. Glipizide 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 34952963-8 2022 In subjects with CYP2C9*1/*3 and CYP2C9*1/*13 genotypes, the mean AUC0- were increased by 44.8% and 58.2%, respectively (both P < 0.001), compared to those of subjects with CYP2C9*1/*1 genotype, while effects of glipizide on plasma glucose and insulin levels were not significantly different between CYP2C9 genotype groups. Glipizide 213-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 34952963-8 2022 In subjects with CYP2C9*1/*3 and CYP2C9*1/*13 genotypes, the mean AUC0- were increased by 44.8% and 58.2%, respectively (both P < 0.001), compared to those of subjects with CYP2C9*1/*1 genotype, while effects of glipizide on plasma glucose and insulin levels were not significantly different between CYP2C9 genotype groups. Glucose 233-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 34952963-9 2022 In conclusion, individuals carrying the defective CYP2C9*3 and CYP2C9*13 alleles have markedly elevated plasma concentrations of glipizide compared with CYP2C9*1/*1 wild-type. Glipizide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 34952963-9 2022 In conclusion, individuals carrying the defective CYP2C9*3 and CYP2C9*13 alleles have markedly elevated plasma concentrations of glipizide compared with CYP2C9*1/*1 wild-type. Glipizide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 34807366-2 2021 Cytochrome P450 (CYP) 2C9 and CYP3A4 are major and minor enzymes involved in the metabolism of meloxicam. Meloxicam 95-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 34807366-3 2021 Impaired enzyme activity of CYP2C9 variants increases the plasma exposures of meloxicam and the risk of adverse events. Meloxicam 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 34807366-12 2021 This study is expected to contribute to reducing the risk of adverse events of meloxicam through optimization of meloxicam dosing in different CYP2C9 genotypes. Meloxicam 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 34817825-0 2021 Physiologically based pharmacokinetic modeling of candesartan related to CYP2C9 genetic polymorphism in adult and pediatric patients. candesartan 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 34817825-3 2021 Candesartan is metabolized by cytochrome P450 2C9 (CYP2C9) enzyme or uridine diphosphate glucurinosyltransferase 1A3, or excreted in an unchanged form through urine, biliary tract and feces. candesartan 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-49 34817825-3 2021 Candesartan is metabolized by cytochrome P450 2C9 (CYP2C9) enzyme or uridine diphosphate glucurinosyltransferase 1A3, or excreted in an unchanged form through urine, biliary tract and feces. candesartan 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 34505930-12 2021 CONCLUSION: Ruxolitinib exposure is increased in GvHD patients in comparison to myelofibrosis patients due to reduced clearance and comedication with CYP3A4 or CYP2C9 inhibitors. ruxolitinib 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 33915070-8 2021 For the inhibition of CYP2C9 and 2E1, pogostone acted as a competitive inhibitor with the Ki value of 6.46 and 7.67 muM and was not affected by the incubation time. Pogostone 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-36 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Cannabinoids 24-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Dronabinol 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 34493602-8 2021 These data suggest that cannabinoids and major THC metabolites are able to inhibit the activities of multiple CYP enzymes, and basic static modelling of these data suggest the possibility of pharmacokinetic interactions between these cannabinoids and xenobiotics extensively metabolized by CYP2B6, CYP2C9 and CYP2D6. Cannabinoids 234-246 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 298-304 34493602-9 2021 Significance Statement Major cannabinoids and their metabolites found in the plasma of cannabis users inhibit several CYP enzymes, including CYP2B6, CYP2C9, and CYP2D6. Cannabinoids 29-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 34149005-2 2021 SMX is metabolized by N-acetyltransferase (NAT) and cytochrome P450 2C9 (CYP2C9) to nontoxic or toxic intermediates. Sulfamethoxazole 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-71 34149005-2 2021 SMX is metabolized by N-acetyltransferase (NAT) and cytochrome P450 2C9 (CYP2C9) to nontoxic or toxic intermediates. Sulfamethoxazole 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 34382722-2 2021 Genetic variation in CYP2C9 and VKORC1 has been the traditional focus in evaluating warfarin dose variability, with little focus on other genes. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 34382722-3 2021 OBJECTIVE: We set out to evaluate 27 single nucleotide polymorphisms (SNPs) in the CYP2C cluster loci and 8 genes (VKORC1, ABCB1, CYP2C9, CYP2C19, CYP2C8, CYP1A2, CYP3A4, and CYP3A5) involved in pharmacokinetics of warfarin. Warfarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 34382722-6 2021 RESULTS: Among black Africans, the SNPs CYP2C rs12777823G>A, CYP2C9 c.449G>A (*8), CYP2C9 c.1003C>T (*11) and CYP2C8 c.805A>T (*2) were significantly associated with warfarin maintenance dose. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34382722-6 2021 RESULTS: Among black Africans, the SNPs CYP2C rs12777823G>A, CYP2C9 c.449G>A (*8), CYP2C9 c.1003C>T (*11) and CYP2C8 c.805A>T (*2) were significantly associated with warfarin maintenance dose. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34382722-8 2021 The presence of CYP2C8*2 and CYP3A5*6 alleles was associated with increased mean warfarin maintenance dose, whereas CYP2C9*8 allele was associated with reduced warfarin maintenance dose. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 34729928-11 2022 The genetic variants within VKORC1 rs9934438 and CYP2C9 rs4086116 explained 39% and 27% of the variability in the weekly warfarin dose requirement in the Qatari and Egyptians, respectively. Warfarin 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 34727989-12 2021 CONCLUSIONS: Based on the clinical course and literature review, we believe that hypoglycemia in the present case was due to a drug interaction, caused by inhibition of CYP2C9 by bucolome and competitive inhibition of CYP2C9 by warfarin, which affected the pharmacokinetics of glimepiride. Warfarin 228-236 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 34727989-12 2021 CONCLUSIONS: Based on the clinical course and literature review, we believe that hypoglycemia in the present case was due to a drug interaction, caused by inhibition of CYP2C9 by bucolome and competitive inhibition of CYP2C9 by warfarin, which affected the pharmacokinetics of glimepiride. glimepiride 277-288 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 34727989-12 2021 CONCLUSIONS: Based on the clinical course and literature review, we believe that hypoglycemia in the present case was due to a drug interaction, caused by inhibition of CYP2C9 by bucolome and competitive inhibition of CYP2C9 by warfarin, which affected the pharmacokinetics of glimepiride. glimepiride 277-288 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 34729928-13 2022 It confirms the importance of VKORC1 and CYP2C9 variants in warfarin dose variability among patients from the MENA region. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 34721047-11 2021 In contrast, ticagrelor-O-glucuronide weakly inhibited CYP2B6, CYP2C9 and CYP2C19 activity with apparent IC50 values of 45.0, 20.0 and 18.8 muM, respectively. ticagrelor-o-glucuronide 13-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 34357562-5 2021 The reduced function alleles CYP2C9*2 and CYP2C9*3, and VKORC1 rs9923231 (c.-1639G>A) are associated with increased sensitivity to warfarin and a low dose requirement to prevent bleeding episodes, whereas CYP4F2 rs2108622 (p.Val433Met) carriers have higher dose requirements (warfarin resistance). Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 34357562-5 2021 The reduced function alleles CYP2C9*2 and CYP2C9*3, and VKORC1 rs9923231 (c.-1639G>A) are associated with increased sensitivity to warfarin and a low dose requirement to prevent bleeding episodes, whereas CYP4F2 rs2108622 (p.Val433Met) carriers have higher dose requirements (warfarin resistance). Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 34357562-5 2021 The reduced function alleles CYP2C9*2 and CYP2C9*3, and VKORC1 rs9923231 (c.-1639G>A) are associated with increased sensitivity to warfarin and a low dose requirement to prevent bleeding episodes, whereas CYP4F2 rs2108622 (p.Val433Met) carriers have higher dose requirements (warfarin resistance). Warfarin 276-284 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 34693729-2 2021 Nala PGx Core is a multi-gene qPCR-based panel of 20 allele variants, comprising 18 SNPs and two CYP2D6 copy number markers across four pharmacogenes - CYP2C9, CYP2C19, CYP2D6 and SLCO1B1. N-acetylleucinamide 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 34498315-2 2021 METHODS: A MassARRAY approach was used to detect single nucleotide polymorphism (SNP) loci in the URAT1 and CYP2C9 genes (16 and 2 loci, respectively) in 111 patients with hypertension and hyperuricemia taking losartan and in 121 healthy controls. Losartan 210-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 34330718-11 2021 Significance Statement This study demonstrates that both CYP2C19 and CYP2C9 are involved in CBD metabolism to the active metabolite 7-OH-CBD, and CYP3A4 is a major contributor to CBD metabolism through pathways other than 7-hydroxylation. 7-Hydroxycannabidiol 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 34330718-12 2021 7-OH-CBD formation was associated with human liver microsomal CYP2C19 activity, but not CYP2C19 genotype, and CYP2C9 was found to contribute significantly to 7-OH-CBD generation. 7-Hydroxycannabidiol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 34330718-12 2021 7-OH-CBD formation was associated with human liver microsomal CYP2C19 activity, but not CYP2C19 genotype, and CYP2C9 was found to contribute significantly to 7-OH-CBD generation. 7-Hydroxycannabidiol 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 34330718-6 2021 The results from reaction phenotyping experiments with recombinant CYP enzymes and CYP-selective chemical inhibitors indicated that both CYP2C19 and CYP2C9 are capable of CBD metabolism to 7-OH-CBD. Cannabidiol 171-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 34330718-6 2021 The results from reaction phenotyping experiments with recombinant CYP enzymes and CYP-selective chemical inhibitors indicated that both CYP2C19 and CYP2C9 are capable of CBD metabolism to 7-OH-CBD. 7-Hydroxycannabidiol 189-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 34330718-9 2021 In a subset of single-donor human liver microsomes with moderate to low CYP2C19 activity, CYP2C9 inhibition significantly reduced 7-OH-CBD formation, suggesting that CYP2C9 may play a greater role in CBD 7-hydroxylation than previously thought. 7-Hydroxycannabidiol 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 34330718-9 2021 In a subset of single-donor human liver microsomes with moderate to low CYP2C19 activity, CYP2C9 inhibition significantly reduced 7-OH-CBD formation, suggesting that CYP2C9 may play a greater role in CBD 7-hydroxylation than previously thought. 7-Hydroxycannabidiol 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 34330718-9 2021 In a subset of single-donor human liver microsomes with moderate to low CYP2C19 activity, CYP2C9 inhibition significantly reduced 7-OH-CBD formation, suggesting that CYP2C9 may play a greater role in CBD 7-hydroxylation than previously thought. Cannabidiol 200-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 34330718-9 2021 In a subset of single-donor human liver microsomes with moderate to low CYP2C19 activity, CYP2C9 inhibition significantly reduced 7-OH-CBD formation, suggesting that CYP2C9 may play a greater role in CBD 7-hydroxylation than previously thought. Cannabidiol 200-203 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 34330718-10 2021 Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD. Cannabidiol 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 34330718-10 2021 Collectively, these data indicate that both CYP2C19 and CYP2C9 are important contributors in CBD metabolism to the active metabolite 7-OH-CBD. 7-Hydroxycannabidiol 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 34330718-11 2021 Significance Statement This study demonstrates that both CYP2C19 and CYP2C9 are involved in CBD metabolism to the active metabolite 7-OH-CBD, and CYP3A4 is a major contributor to CBD metabolism through pathways other than 7-hydroxylation. Cannabidiol 92-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 34429480-1 2021 CYP2C9, one of the most abundant hepatic cytochrome P450 enzymes, is involved in metabolism of 15-20% of clinically important drugs (warfarin, sulfonylureas, phenytoin, non-steroid anti-inflammatory drugs). Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34106453-1 2021 BACKGROUND: Several studies optimized the warfarin dose based on CYP2C9*2, CYP2C9*3, VKORC1 -1639 G > A, CYP4F2 V433M. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 34106453-1 2021 BACKGROUND: Several studies optimized the warfarin dose based on CYP2C9*2, CYP2C9*3, VKORC1 -1639 G > A, CYP4F2 V433M. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 34680932-1 2021 The genotypes of vitamin K epoxide reductase complex 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) can influence therapeutic warfarin doses. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-87 34680932-1 2021 The genotypes of vitamin K epoxide reductase complex 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) can influence therapeutic warfarin doses. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 34900803-0 2021 A pharmacogenetic pilot study of CYP2C9 common genetic variant and sulfonylureas therapeutic response in type 2 diabetes mellitus patients. Sulfonylurea Compounds 67-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 34900803-2 2021 Sulfonylureas (SFUs) are the most widely used among the oral antidiabetic drugs that are highly metabolized by cytochrome P450 family 2 subfamily C member 9 (CYP2C9). Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-156 34900803-2 2021 Sulfonylureas (SFUs) are the most widely used among the oral antidiabetic drugs that are highly metabolized by cytochrome P450 family 2 subfamily C member 9 (CYP2C9). Sulfonylurea Compounds 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 34900803-2 2021 Sulfonylureas (SFUs) are the most widely used among the oral antidiabetic drugs that are highly metabolized by cytochrome P450 family 2 subfamily C member 9 (CYP2C9). Sulfonylurea Compounds 15-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-156 34900803-2 2021 Sulfonylureas (SFUs) are the most widely used among the oral antidiabetic drugs that are highly metabolized by cytochrome P450 family 2 subfamily C member 9 (CYP2C9). Sulfonylurea Compounds 15-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 34314704-2 2021 In particular, the anti-coagulant warfarin is prescribed to over 15 million people annually and polymorphisms in CYP2C9 can affect individual drug response and lead to an increased risk of hemorrhage. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 34273250-0 2021 Prediction of the drug-drug interaction potential of the alpha-acid glycoprotein bound, CYP3A4/CYP2C9 metabolized oncology drug, erdafitinib. erdafitinib 129-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 34273250-2 2021 Erdafitinib binds preferentially to alpha1-acid glycoprotein (AGP), and is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. erdafitinib 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-125 34273250-3 2021 This article describes a physiologically based pharmacokinetic (PBPK) model for erdafitinib to assess the drug-drug interaction (DDI) potential of CYP3A4 and CYP2C9 inhibitors, and CYP3A4/CYP2C9 inducers on erdafitinib PK in cancer patients exhibiting higher AGP levels and in population with different CYP2C9 genotypes. erdafitinib 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 34273250-3 2021 This article describes a physiologically based pharmacokinetic (PBPK) model for erdafitinib to assess the drug-drug interaction (DDI) potential of CYP3A4 and CYP2C9 inhibitors, and CYP3A4/CYP2C9 inducers on erdafitinib PK in cancer patients exhibiting higher AGP levels and in population with different CYP2C9 genotypes. erdafitinib 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 34273250-3 2021 This article describes a physiologically based pharmacokinetic (PBPK) model for erdafitinib to assess the drug-drug interaction (DDI) potential of CYP3A4 and CYP2C9 inhibitors, and CYP3A4/CYP2C9 inducers on erdafitinib PK in cancer patients exhibiting higher AGP levels and in population with different CYP2C9 genotypes. erdafitinib 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 303-309 34273250-3 2021 This article describes a physiologically based pharmacokinetic (PBPK) model for erdafitinib to assess the drug-drug interaction (DDI) potential of CYP3A4 and CYP2C9 inhibitors, and CYP3A4/CYP2C9 inducers on erdafitinib PK in cancer patients exhibiting higher AGP levels and in population with different CYP2C9 genotypes. erdafitinib 207-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 34273250-6 2021 Erdafitinib clearance in the PBPK model consisted of multiple pathways (CYP2C9/3A4, renal, intestinal; additional hepatic clearance), making the compound less susceptible to DDIs. erdafitinib 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-82 34273250-7 2021 In poor metabolizing CYP2C9 populations carrying the CYP2C9*3/*3 genotype, simulations shown clinically relevant increase in erdafitinib plasma concentrations. erdafitinib 125-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 34273250-7 2021 In poor metabolizing CYP2C9 populations carrying the CYP2C9*3/*3 genotype, simulations shown clinically relevant increase in erdafitinib plasma concentrations. erdafitinib 125-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 34664206-5 2021 The processes implemented to describe the rosuvastatin pharmacokinetics and DDIs are active uptake by OATP2B1, OATP1B1/OATP1B3 and OAT3, active efflux by BCRP and Pgp, metabolism by CYP2C9 and passive glomerular filtration. Rosuvastatin Calcium 42-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 34638547-5 2021 The X-ray crystal structure of CYP2C9*8, which represents an amino acid variation from arginine to histidine at position 150 (R150H), was solved in complex with losartan. Losartan 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 34638547-6 2021 The overall conformation of the CYP2C9*8-losartan complex was similar to the previously solved complex with wild type (WT) protein, but it differs in the occupancy of losartan. 8-losartan 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 34638547-6 2021 The overall conformation of the CYP2C9*8-losartan complex was similar to the previously solved complex with wild type (WT) protein, but it differs in the occupancy of losartan. Losartan 167-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 34638547-10 2021 Interestingly, the CYP2C9*8 interaction with losartan was not as weak as the CYP2C9*3 variant, which showed up to three-fold weaker average dissociation constant compared to the WT. Losartan 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 34638547-11 2021 Taken together, the structural and solution characterization yields insights into the similarities and differences of losartan binding to CYP2C9 variants and provides a useful framework for probing the role of amino acid substitution and substrate dependent activity. Losartan 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 34575495-2 2021 Resveratrol enantiomers showed stereoselective inhibition of CYP2C9, CYP3A, and UGT1A1. Resveratrol 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34109627-2 2021 Genetic variation within the CYP2C9 gene locus impacts the metabolism or bioactivation of many clinically important drugs including NSAIDs, phenytoin, anti-diabetic agents and angiotensin receptor blocker. Phenytoin 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 34109627-3 2021 Variable CYP2C9 activity is of particular importance regarding efficacy and safety of warfarin and siponimod as indicated in their package inserts. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 34109627-3 2021 Variable CYP2C9 activity is of particular importance regarding efficacy and safety of warfarin and siponimod as indicated in their package inserts. siponimod 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 34429480-1 2021 CYP2C9, one of the most abundant hepatic cytochrome P450 enzymes, is involved in metabolism of 15-20% of clinically important drugs (warfarin, sulfonylureas, phenytoin, non-steroid anti-inflammatory drugs). Sulfonylurea Compounds 143-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34429480-1 2021 CYP2C9, one of the most abundant hepatic cytochrome P450 enzymes, is involved in metabolism of 15-20% of clinically important drugs (warfarin, sulfonylureas, phenytoin, non-steroid anti-inflammatory drugs). Phenytoin 158-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34429480-1 2021 CYP2C9, one of the most abundant hepatic cytochrome P450 enzymes, is involved in metabolism of 15-20% of clinically important drugs (warfarin, sulfonylureas, phenytoin, non-steroid anti-inflammatory drugs). Steroids 173-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34429480-4 2021 The presence of CYP2C9*3 allele was associated with CYP2C9 functional impairment, and CYP2C9*2 influenced tolbutamide 4"-hydroxylase activity only in subjects with two polymorphic alleles, whereas the contribution of CYP2C8*3 was not confirmed. Tolbutamide 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 34429480-5 2021 In addition to CYP2C9 genetic polymorphisms, non-genetic factors (co-medication with CYP2C9-specific inhibitors/inducers and non-specific factors including amoxicillin + clavulanic acid therapy or chronic alcohol consumption) contributed to the prediction of hepatic CYP2C9 activity; however, a CYP2C9 genotype-phenotype mismatch still existed in 32.6% of the subjects. Amoxicillin-Potassium Clavulanate Combination 156-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 267-273 34429480-5 2021 In addition to CYP2C9 genetic polymorphisms, non-genetic factors (co-medication with CYP2C9-specific inhibitors/inducers and non-specific factors including amoxicillin + clavulanic acid therapy or chronic alcohol consumption) contributed to the prediction of hepatic CYP2C9 activity; however, a CYP2C9 genotype-phenotype mismatch still existed in 32.6% of the subjects. Alcohols 205-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 267-273 34194474-11 2021 The CYP2C9 alleles, * 2 (rs1799853C>T) and * 3 (rs1057910A>C) that are predictive of low enzyme activity were more common in subjects who experienced hypoglycemia after treatment with sulfonylureas. Sulfonylurea Compounds 184-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 34242938-7 2021 The final PBPK/PD model of warfarin enantiomers was verified and validated by different dosing regimens, ethnicities and genetic polymorphisms, and used to perform DDI simulations between warfarin racemate and sorafenib among general populations and sub-populations with various CYP2C9 and VKORC1 genotypes. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 279-285 34242938-7 2021 The final PBPK/PD model of warfarin enantiomers was verified and validated by different dosing regimens, ethnicities and genetic polymorphisms, and used to perform DDI simulations between warfarin racemate and sorafenib among general populations and sub-populations with various CYP2C9 and VKORC1 genotypes. Sorafenib 210-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 279-285 34074714-4 2021 The IC50 for inhibition of CYP2C8, CYP2C9, and CYP3A4/5 for verinurad was {greater than or equal to}14.5 microM, and Iu,max/IC50 was <0.02 at the anticipated therapeutic Cmax and therefore was not considered a DDI risk. verinurad 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 34246203-4 2021 The aims of the present study are to assess the possible association of CYP2D6 and CYP2C9 polymorphisms with the efficacy and safety of tramadol and ketorolac in postoperative pain. Tramadol 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34246203-4 2021 The aims of the present study are to assess the possible association of CYP2D6 and CYP2C9 polymorphisms with the efficacy and safety of tramadol and ketorolac in postoperative pain. Ketorolac 149-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34246203-15 2021 CONCLUSIONS: CYP2D6 and CYP2C9 can predict analgesic effectiveness of tramadol and ketorolac. Tramadol 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 34246203-15 2021 CONCLUSIONS: CYP2D6 and CYP2C9 can predict analgesic effectiveness of tramadol and ketorolac. Ketorolac 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 34246203-16 2021 CYP2C9 can predict the risk of gastrointestinal bleeding, including those hidden to ketorolac. Ketorolac 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34304363-0 2021 Physiologically based pharmacokinetic (PBPK) modeling for prediction of celecoxib pharmacokinetics according to CYP2C9 genetic polymorphism. Celecoxib 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 34304363-3 2021 Many studies reported that CYP2C9 genetic polymorphism has significant effects on the pharmacokinetics of celecoxib and the occurrence of adverse drug reactions. Celecoxib 106-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 34304363-4 2021 The aim of this study was to develop a physiologically based pharmacokinetic (PBPK) model of celecoxib according to CYP2C9 genetic polymorphism for personalized pharmacotherapy. Celecoxib 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 34304363-6 2021 Based on the conducted pharmacokinetic study and a previous pharmacokinetic study involving subjects with CYP2C9*1/*13 and CYP2C9*3/*3 genotype, PBPK model for celecoxib was developed. Celecoxib 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 34304363-6 2021 Based on the conducted pharmacokinetic study and a previous pharmacokinetic study involving subjects with CYP2C9*1/*13 and CYP2C9*3/*3 genotype, PBPK model for celecoxib was developed. Celecoxib 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 34304363-9 2021 As a result, the developed PBPK model of celecoxib successfully described the pharmacokinetics of each CYP2C9 genotype group and its predicted values were within the acceptance criterion. Celecoxib 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 34304363-11 2021 This study demonstrates the possibility of determining the appropriate dosage of celecoxib for each individual through the PBPK modeling with CYP2C9 genomic information. Celecoxib 81-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 34143391-5 2021 Lenabasum is a CYP substrate, and the model predicted lenabasum clearance of 51% by CYP2C9, 37% by CYP2C8, and 12% by CYP3A4. lenabasum 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 34143391-5 2021 Lenabasum is a CYP substrate, and the model predicted lenabasum clearance of 51% by CYP2C9, 37% by CYP2C8, and 12% by CYP3A4. lenabasum 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 34060344-0 2021 A meta-analysis of effects of CYP2C9 and CYP2C19 polymorphisms on phenytoin pharmacokinetic parameters. Phenytoin 66-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 34060344-1 2021 Aim: Phenytoin is metabolized through CYP2C9 and CYP2C19. Phenytoin 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 34060344-8 2021 Conclusion: The dosage regimen for patients in the CYP2C9IM group to achieve phenytoin therapeutic levels was 2.1-3.4 mg/kg/day. Phenytoin 77-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 34210056-0 2021 Influence of CYP2C9 Genetic Polymorphisms on the Pharmacokinetics of Losartan and Its Active Metabolite E-3174: A Systematic Review and Meta-Analysis. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 34210056-0 2021 Influence of CYP2C9 Genetic Polymorphisms on the Pharmacokinetics of Losartan and Its Active Metabolite E-3174: A Systematic Review and Meta-Analysis. losartan carboxylic acid 104-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 34210056-1 2021 This study aimed to investigate the influence of CYP2C9 genetic polymorphisms on the pharmacokinetics of losartan and its active metabolite, E-3174, through a systematic review and meta-analysis. Losartan 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 34210056-5 2021 We found that healthy volunteers with CYP2C9*2 or *3 carriers had higher area under the curve (AUC0- ) of losartan (mean difference (MD) 0.17 mug h/mL; 95% confidence intervals (CI): 0.04, 0.29) and lower AUC0- of E-3174 (MD -0.35 mug h/mL; 95% CI: -0.62, -0.08) than those with CYP2C9*1/*1. Losartan 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 34210056-7 2021 For half-life, subjects with CYP2C9*2 or *3 carriers had longer half-lives of losartan and E-3174 than those with CYP2C9*1/*1 (MD 0.47 h; 95% CI: 0.32, 0.61 and MD 0.68 h; 95% CI: 0.44, 0.92, respectively). Losartan 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 34210056-7 2021 For half-life, subjects with CYP2C9*2 or *3 carriers had longer half-lives of losartan and E-3174 than those with CYP2C9*1/*1 (MD 0.47 h; 95% CI: 0.32, 0.61 and MD 0.68 h; 95% CI: 0.44, 0.92, respectively). losartan carboxylic acid 91-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 34210056-8 2021 This meta-analysis suggests that the pharmacokinetics of losartan and E-3174 are associated with the CYP2C9 polymorphisms. Losartan 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 34210056-8 2021 This meta-analysis suggests that the pharmacokinetics of losartan and E-3174 are associated with the CYP2C9 polymorphisms. losartan carboxylic acid 70-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 34082641-5 2021 The inhibition of CYP2C9 and 2D was not affected by the incubation time but was found to be competitive with the Ki values of 6.7 and 6.6 muM, respectively.The inhibitory effect of 4-O-galloylalbiflorin on the activity of CYP3A, 2C9, and 2D implying the potential drug-drug interaction between 4-O-galloylalbiflorin and the drugs metabolized by these CYP450s. 4-O-galloylalbiflorin 181-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 34082641-5 2021 The inhibition of CYP2C9 and 2D was not affected by the incubation time but was found to be competitive with the Ki values of 6.7 and 6.6 muM, respectively.The inhibitory effect of 4-O-galloylalbiflorin on the activity of CYP3A, 2C9, and 2D implying the potential drug-drug interaction between 4-O-galloylalbiflorin and the drugs metabolized by these CYP450s. 4-O-galloylalbiflorin 294-315 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 34228652-0 2021 Impact of VKORC1, CYP2C9, and CYP4F2 Polymorphisms on Optimal Warfarin Dose: Does Ethnicity Matters? Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 34181150-11 2021 CYP2C9-mediated metabolism was inhibited by nearly all the cannabinoids with estimated Ki values of 0.2-3.2 muM. Cannabinoids 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34181150-14 2021 CYP2C9-mediated metabolism was inhibited by cannabinoids at clinically relevant concentrations. Cannabinoids 44-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34104863-8 2021 The combination of azithromycin intake, a known inhibitor of CYP2C9, with the presence of CYP2C9*2 and -1639G>A VKORC1, two variants associated with warfarin hypersensitivity, have likely contributed to explain the warfarin overdose and the difficulty to reverse warfarin effect in this patient. Warfarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34104863-8 2021 The combination of azithromycin intake, a known inhibitor of CYP2C9, with the presence of CYP2C9*2 and -1639G>A VKORC1, two variants associated with warfarin hypersensitivity, have likely contributed to explain the warfarin overdose and the difficulty to reverse warfarin effect in this patient. Azithromycin 19-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34104863-8 2021 The combination of azithromycin intake, a known inhibitor of CYP2C9, with the presence of CYP2C9*2 and -1639G>A VKORC1, two variants associated with warfarin hypersensitivity, have likely contributed to explain the warfarin overdose and the difficulty to reverse warfarin effect in this patient. Azithromycin 19-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 34116570-11 2021 Herbacetin inhibited CYP3A4, CYP2B6, CYP2C9, and CYP2E1 in a mixed manner, but non-competitively blocked CYP2D6. herbacetin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 34104863-8 2021 The combination of azithromycin intake, a known inhibitor of CYP2C9, with the presence of CYP2C9*2 and -1639G>A VKORC1, two variants associated with warfarin hypersensitivity, have likely contributed to explain the warfarin overdose and the difficulty to reverse warfarin effect in this patient. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34104863-8 2021 The combination of azithromycin intake, a known inhibitor of CYP2C9, with the presence of CYP2C9*2 and -1639G>A VKORC1, two variants associated with warfarin hypersensitivity, have likely contributed to explain the warfarin overdose and the difficulty to reverse warfarin effect in this patient. Warfarin 263-271 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 34092821-4 2021 Objective: The objective of this study was to investigate the effect of polymorphisms of the CYP3A4, CYP2C9, CYP3A5, ABCB1, CYP2C19, SCL6A4, and 5-HTR2A genes on the concentration/dose indicator of mirtazapine and on the CYP3A expression level obtained by measuring the miR-27b plasma concentration levels in patients suffering from a recurrent depressive disorder. Mirtazapine 198-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 34199712-10 2021 CYP3A4 and CYP2C9 were also responsible for the metabolism of tamoxifen. Tamoxifen 62-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 34199712-11 2021 CYP2C9 especially impacted the hydroxylation to 4-OHtam, and this involved the OATP1B1 (SLCO1B1) transporter. 4-ohtam 48-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34104133-2 2021 Objectives: To identify occurrence of polymorphisms of the CYP2C9 and VKORC1 genes in patients taking warfarin and relate these profiles to their medication dosages and the Time in Therapeutic Range (TTR). Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 34471061-4 2021 On comparing the results of ES with those of estradiol (E2), it was found that CYP2C8 was about 2.5 times higher and CYP2C9 was about 3 times higher, and ES was more likely to be a substrate for the 2-hydroxylation reaction by both CYPs. estradiol 17-sulfate 28-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 34164480-0 2021 The association of adjusted plasma valproic acid concentration with CYP2C9 gene polymorphism in patients with epilepsy: a systematic review and meta-analysis. Valproic Acid 35-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 34164480-2 2021 Although there is increasing evidence to suggest that CYP2C9 gene polymorphisms are associated with interindividual variability of VPA metabolism, the results are debatable. Valproic Acid 131-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 34164480-3 2021 Therefore, in the present study, we conducted a meta-analysis to evaluate the correlation between CYP2C9 gene polymorphisms and adjusted plasma VPA concentration. Valproic Acid 144-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 34164480-6 2021 We calculated 95% confidence intervals (CIs) and mean differences (MDs) to assess the strength of the relationship of CYP2C9 gene polymorphisms with adjusted plasma VPA concentration. Valproic Acid 165-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 34164480-8 2021 The pooled analysis showed that the CYP2C9 A1075C (AA vs. AC) polymorphism was related to the adjusted plasma concentration of VPA (P=0.02, I2= 82%). Valproic Acid 127-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 34164480-12 2021 A reduced VPA dosage may be recommendable for children, particularly Asian children, who are CYP2C9 A1075C AC carriers. Valproic Acid 10-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 34164480-13 2021 Further studies could provide high-quality evidence to confirm the correlation between VPA pharmacokinetics and CYP2C9 A1075C polymorphisms. Valproic Acid 87-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 34471061-4 2021 On comparing the results of ES with those of estradiol (E2), it was found that CYP2C8 was about 2.5 times higher and CYP2C9 was about 3 times higher, and ES was more likely to be a substrate for the 2-hydroxylation reaction by both CYPs. Estradiol 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 34471061-4 2021 On comparing the results of ES with those of estradiol (E2), it was found that CYP2C8 was about 2.5 times higher and CYP2C9 was about 3 times higher, and ES was more likely to be a substrate for the 2-hydroxylation reaction by both CYPs. Estradiol 56-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Diclofenac 212-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34272716-0 2021 Case Study 9: Probe-Dependent Binding Explains Lack of CYP2C9 Inactivation by 1-Aminobenzotriazole (ABT). 1-aminobenzotriazole 78-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Heme 268-272 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34272716-0 2021 Case Study 9: Probe-Dependent Binding Explains Lack of CYP2C9 Inactivation by 1-Aminobenzotriazole (ABT). 1-aminobenzotriazole 100-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 34272716-7 2021 ABT preferentially binds in the distal warfarin-binding pocket, supporting that diclofenac is minimally deterred from access to the CYP2C9 active site in the presence of ABT, thus resulting in minimal inactivation. Diclofenac 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Diclofenac 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Diclofenac 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 34272716-7 2021 ABT preferentially binds in the distal warfarin-binding pocket, supporting that diclofenac is minimally deterred from access to the CYP2C9 active site in the presence of ABT, thus resulting in minimal inactivation. 1-aminobenzotriazole 170-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Diclofenac 156-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 34272716-8 2021 Simultaneously docking of (S)-warfarin and ABT revealed that (S)-warfarin outcompetes ABT for the distal binding site and results in the binding of ABT to the CYP2C9 active site, supporting the observations of potent inactivation of CYP2C9 when (S)-warfarin is the probe substrate.These results highlight that probe selection is crucial when evaluating CYP inhibition potential, and it is recommended that multiple probes be utilized for CYP2C9, similar to the approach routinely employed for CYP3A4. Warfarin 61-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). Warfarin 181-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 34272716-8 2021 Simultaneously docking of (S)-warfarin and ABT revealed that (S)-warfarin outcompetes ABT for the distal binding site and results in the binding of ABT to the CYP2C9 active site, supporting the observations of potent inactivation of CYP2C9 when (S)-warfarin is the probe substrate.These results highlight that probe selection is crucial when evaluating CYP inhibition potential, and it is recommended that multiple probes be utilized for CYP2C9, similar to the approach routinely employed for CYP3A4. 1-aminobenzotriazole 148-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 458-478 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 34272716-8 2021 Simultaneously docking of (S)-warfarin and ABT revealed that (S)-warfarin outcompetes ABT for the distal binding site and results in the binding of ABT to the CYP2C9 active site, supporting the observations of potent inactivation of CYP2C9 when (S)-warfarin is the probe substrate.These results highlight that probe selection is crucial when evaluating CYP inhibition potential, and it is recommended that multiple probes be utilized for CYP2C9, similar to the approach routinely employed for CYP3A4. Warfarin 245-257 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 458-478 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 458-478 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 423-429 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 480-483 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 480-483 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 295-301 34272716-3 2021 In evaluating a time-saving assay using diclofenac as the CYP2C9 probe substrate, a discrepancy was observed in which minimal inhibition was detected using diclofenac whereas using (S)-warfarin resulted in potent inhibition, supporting the presence of dual-binding sites in the relatively large CYP2C9 active site cavity.These observations provided further insights into explaining the reported ineffective inactivation of CYP2C9 for the pan-CYP inactivator 1-aminobenzotriazole (ABT). 1-aminobenzotriazole 480-483 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 423-429 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. 1-aminobenzotriazole 118-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. 1-aminobenzotriazole 118-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 229-235 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. Warfarin 168-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. Warfarin 168-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 229-235 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. 1-aminobenzotriazole 250-253 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. 1-aminobenzotriazole 250-253 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 229-235 34272716-4 2021 Mechanistic reversible and time-dependent inhibition experiments revealed that the ineffective CYP2C9 inactivation by ABT was also probe-dependent, with utilization of (S)-warfarin as the probe substrate resulting in more potent CYP2C9 inhibition by ABT compared to diclofenac. Diclofenac 266-276 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 34272716-5 2021 Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. Warfarin 12-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 34272716-5 2021 Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. 1-aminobenzotriazole 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 34272716-5 2021 Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. Diclofenac 101-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 34272716-5 2021 Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. 1-aminobenzotriazole 168-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 34272716-5 2021 Addition of (S)-warfarin to the reversible and time-dependent inhibition experiments between ABT and diclofenac resulted in an attenuation of the inhibitory effects of ABT on CYP2C9-mediated diclofenac metabolism. Diclofenac 191-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 34802403-0 2021 Influence of CYP2C9 Polymorphisms on Plasma Concentration of Warfarin and 7-Hydroxy Warfarin in South Indian Patients. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 34802403-1 2021 BACKGROUND: Warfarin is primarily metabolized by cytochrome P450 2C9 (CYP2C9), which is encoded by the CYP2C9 gene. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-68 34802403-1 2021 BACKGROUND: Warfarin is primarily metabolized by cytochrome P450 2C9 (CYP2C9), which is encoded by the CYP2C9 gene. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 34802403-1 2021 BACKGROUND: Warfarin is primarily metabolized by cytochrome P450 2C9 (CYP2C9), which is encoded by the CYP2C9 gene. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 34802403-2 2021 CYP2C9*2 and CYP2C9*3 variants significantly influence warfarin metabolism and subsequently the required dose of warfarin. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34802403-2 2021 CYP2C9*2 and CYP2C9*3 variants significantly influence warfarin metabolism and subsequently the required dose of warfarin. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 34802403-2 2021 CYP2C9*2 and CYP2C9*3 variants significantly influence warfarin metabolism and subsequently the required dose of warfarin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 34802403-2 2021 CYP2C9*2 and CYP2C9*3 variants significantly influence warfarin metabolism and subsequently the required dose of warfarin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34272716-9 2021 Further, utilization of ABT as a pan-inhibitor of CYP activity for investigational compounds, both in vitro and in vivo, should be accompanied with the understanding that residual CYP-mediated oxidative metabolism could potentially be observed for CYP2C9 substrates and should not necessarily be attributed to non-P450-mediated metabolism. 1-aminobenzotriazole 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 248-254 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Diclofenac 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34802403-3 2021 OBJECTIVE: The current retrospective study was aimed to determine the influence of CYP2C9 variants on warfarin metabolic ratio (MR, warfarin/7-hydroxy warfarin) and warfarin maintenance therapy in 210 patients (mean age 44.6+-11.6 (SD) years; male to female ratio 81:129). Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34802403-3 2021 OBJECTIVE: The current retrospective study was aimed to determine the influence of CYP2C9 variants on warfarin metabolic ratio (MR, warfarin/7-hydroxy warfarin) and warfarin maintenance therapy in 210 patients (mean age 44.6+-11.6 (SD) years; male to female ratio 81:129). Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34802403-3 2021 OBJECTIVE: The current retrospective study was aimed to determine the influence of CYP2C9 variants on warfarin metabolic ratio (MR, warfarin/7-hydroxy warfarin) and warfarin maintenance therapy in 210 patients (mean age 44.6+-11.6 (SD) years; male to female ratio 81:129). 7-hydroxywarfarin 141-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 34802403-9 2021 Patients carrying other CYP2C9 variants required 39% lower warfarin maintenance dose (3.3+-1.2 mg) than CYP2C9*1*1 carrier (4.9+-1.8 mg), (p<0.0001). Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 34802403-12 2021 CONCLUSION: The present study results provide, insights on effect of CYP2C9 genetic polymorphisms on inter-individual variability in warfarin metabolism and emphasizes utility of phenotyping in a setting of genotype-guided dosing of warfarin in the South Indian population. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 34272716-6 2021 Molecular docking studies further confirmed that (S)-warfarin and diclofenac preferentially bind in different regions of the CYP2C9 active site, with (S)-warfarin occupying a distal "warfarin-binding pocket" and diclofenac occupying a binding site close to the active heme moiety. Warfarin 183-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 34900058-6 2021 Results: Ifosfamide levels increased according to the genotype, and patients with the variant rs1799853 in CYP2C9 genotype CC had lower levels of ifosfamide (median = 1.8 mumol/l, Q25 0.9-Q75 4.6) compared with patients with genotype TT + CT (median = 2.8 mumol/l, Q25 1.9-Q75 5.1), p < 0.001. Ifosfamide 146-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 34900058-9 2021 Conclusions: Polymorphisms in CYP2C9 and CYP3A4 genes are associated with high levels of ifosfamide. Ifosfamide 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Rifampin 151-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 35370222-1 2022 We encountered cases in which the anticoagulant effects of warfarin (CYP2C9 substrate) were reversibly attenuated by the concomitant administration of rifampicin or bosentan, which are potent pregnane X receptor (PXR) ligands. Bosentan 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 35168147-5 2022 Flurbiprofen/naproxen and piroxicam are located in the active site and the primary binding site of CYP2C9, respectively. Piroxicam 26-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. Dronabinol 14-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. Dronabinol 14-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. 11-Hydroxytetrahydrocannabinol 18-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. 11-Hydroxytetrahydrocannabinol 18-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 35168147-1 2022 Some non-steroidal anti-inflammatory drugs (NSAIDs) exhibit atypical kinetic behavior when binding together with dapsone in CYP2C9. Dapsone 113-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 35168147-3 2022 Here, molecular docking and molecular dynamics (MD) simulations are performed to explore the cooperative binding of dapsone and three NSAIDs in CYP2C9. Dapsone 116-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. Dronabinol 95-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 35168147-5 2022 Flurbiprofen/naproxen and piroxicam are located in the active site and the primary binding site of CYP2C9, respectively. Flurbiprofen 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 35599347-5 2022 FFA and nFFA were direct in vitro inhibitors of CYP2D6 (IC50 , 4.7 and 16 microM, respectively) but did not substantially inhibit CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A4/5. Fenfluramine 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 35168147-5 2022 Flurbiprofen/naproxen and piroxicam are located in the active site and the primary binding site of CYP2C9, respectively. Naproxen 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 35061161-2 2022 The herbal hepatoprotective medicine, silybin A inhibits cytochrome P450 (CYP) 2C9 and 3A4 enzymes, thus, may interact with the drugs that are substrates of CYP2C9 and 3A4, such as losartan. Silybin 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-90 35061161-2 2022 The herbal hepatoprotective medicine, silybin A inhibits cytochrome P450 (CYP) 2C9 and 3A4 enzymes, thus, may interact with the drugs that are substrates of CYP2C9 and 3A4, such as losartan. Silybin 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 35061161-2 2022 The herbal hepatoprotective medicine, silybin A inhibits cytochrome P450 (CYP) 2C9 and 3A4 enzymes, thus, may interact with the drugs that are substrates of CYP2C9 and 3A4, such as losartan. Losartan 181-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-90 35061161-2 2022 The herbal hepatoprotective medicine, silybin A inhibits cytochrome P450 (CYP) 2C9 and 3A4 enzymes, thus, may interact with the drugs that are substrates of CYP2C9 and 3A4, such as losartan. Losartan 181-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 35061161-4 2022 This study aimed to assess silybin A-losartan interaction in different CYP2C9 genotypes using physiological-based pharmacokinetic (PBPK) model approach. Silybin 27-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 35061161-4 2022 This study aimed to assess silybin A-losartan interaction in different CYP2C9 genotypes using physiological-based pharmacokinetic (PBPK) model approach. Losartan 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 35061161-10 2022 In CYP 2C9*1/*1 genotype, AUC and Cmax of losartan were increased 1.16 and 1.37 folds, respectively falling in a range stipulated for negligible interaction. Losartan 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-10 35061161-11 2022 Increase in AUC and Cmax by 0.873 and 0.294 folds, respectively in CYP2C9*1/*3 after co-administration of silybin A exhibited a minor interaction with losartan. Silybin 106-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 35061161-11 2022 Increase in AUC and Cmax by 0.873 and 0.294 folds, respectively in CYP2C9*1/*3 after co-administration of silybin A exhibited a minor interaction with losartan. Losartan 151-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 35061161-12 2022 However, in individuals with CYP2C9*1/*2 genotype, the losartan"s AUC and Cmax were decreased by 0.01 folds, manifesting a moderate interaction. Losartan 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 35061161-13 2022 Hence, in CYP2C9*1/*1 and CYP2C9*1/*3 genotypes, silybin A is a weak CYP inhibitor for losartan while in CYP2C9*1/*2 genotype, the co-administration of silybin consequents into a moderate pharmacokinetic interaction with losartan. Losartan 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. Dronabinol 95-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 35370140-3 2022 To populate a THC/11-OH-THC PBPK model, we previously characterized the depletion clearance of THC (by CYP2C9) and 11-OH-THC (by UGT, CYP3A, and CYP2C9) in adult human liver microsomes. 11-Hydroxytetrahydrocannabinol 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 35081606-0 2022 Effects of CYP2C19, CYP2C9 and CYP3A4 gene polymorphisms on plasma voriconazole levels in Chinese pediatric patients. Voriconazole 67-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 35081606-2 2022 Previous studies suggested that voriconazole was extensively metabolized by CYP450 enzyme system, including CYP2C19, CYP2C9 and CYP3A4, which contributed to the individual variability of the pharmacokinetic process of voriconazole. Voriconazole 32-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 35081606-2 2022 Previous studies suggested that voriconazole was extensively metabolized by CYP450 enzyme system, including CYP2C19, CYP2C9 and CYP3A4, which contributed to the individual variability of the pharmacokinetic process of voriconazole. Voriconazole 218-230 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 35081606-3 2022 This study aimed to investigate the effects of CYP2C19, CYP2C9 and CYP3A4 gene polymorphisms on plasma voriconazole concentrations in Chinese pediatric patients. Voriconazole 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 35599347-5 2022 FFA and nFFA were direct in vitro inhibitors of CYP2D6 (IC50 , 4.7 and 16 microM, respectively) but did not substantially inhibit CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, or CYP3A4/5. Norfenfluramine 8-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 35583123-0 2022 Methysticin Acts as a Mechanism-Based Inactivator of Cytochrome P450 2C9. methysticin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 35622425-4 2022 DDI for warfarin mainly involve moderate to strong inhibitors/inducers of cytochrome P450 (CYP) 2C9, which is responsible for the elimination of the more potent S-isomer of warfarin. Warfarin 8-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-99 35622425-4 2022 DDI for warfarin mainly involve moderate to strong inhibitors/inducers of cytochrome P450 (CYP) 2C9, which is responsible for the elimination of the more potent S-isomer of warfarin. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-99 35583123-12 2022 Both ortho-quinone and carbene intermediates appeared to be involved in the inactivation of CYP2C9 induced by methysticin. 2-benzoquinone 5-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 35590072-6 2022 For PharmGKB level 1A variants, CYP2C9 status for phenytoin prescription was the only relevant information. Phenytoin 50-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 35583123-3 2022 Methysticin exhibited time-, concentration-, and NADPH-dependent inhibition on CYP2C9 using diclofenac as a probe substrate. methysticin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 35583123-12 2022 Both ortho-quinone and carbene intermediates appeared to be involved in the inactivation of CYP2C9 induced by methysticin. carbene 23-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 35583123-3 2022 Methysticin exhibited time-, concentration-, and NADPH-dependent inhibition on CYP2C9 using diclofenac as a probe substrate. NADP 49-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 35583123-12 2022 Both ortho-quinone and carbene intermediates appeared to be involved in the inactivation of CYP2C9 induced by methysticin. methysticin 110-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 35583123-3 2022 Methysticin exhibited time-, concentration-, and NADPH-dependent inhibition on CYP2C9 using diclofenac as a probe substrate. Diclofenac 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 35583123-4 2022 Approximately 85% of CYP2C9 activity was inhibited by methysticin at 50 muM after a 30 min preincubation with human liver microsomes in the presence of NADPH. methysticin 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 35583123-4 2022 Approximately 85% of CYP2C9 activity was inhibited by methysticin at 50 muM after a 30 min preincubation with human liver microsomes in the presence of NADPH. NADP 152-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 35583123-6 2022 Sulfaphenazole (competitive inhibitor of CYP2C9) displayed a significant protective effect on methysticin-induced CYP2C9 inactivation. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 35583123-6 2022 Sulfaphenazole (competitive inhibitor of CYP2C9) displayed a significant protective effect on methysticin-induced CYP2C9 inactivation. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 35583123-6 2022 Sulfaphenazole (competitive inhibitor of CYP2C9) displayed a significant protective effect on methysticin-induced CYP2C9 inactivation. methysticin 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 35583123-6 2022 Sulfaphenazole (competitive inhibitor of CYP2C9) displayed a significant protective effect on methysticin-induced CYP2C9 inactivation. methysticin 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. carbene 2-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. carbene 2-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. methysticin 56-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. methysticin 56-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. k3fe 99-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 35583123-8 2022 A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. k3fe 99-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 35583123-9 2022 A methysticin-derived ortho-quinone intermediate dependent on NADPH was trapped by GSH, and this intermediate was believed to be involved in CYP2C9 inactivation. methysticin 2-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 35583123-9 2022 A methysticin-derived ortho-quinone intermediate dependent on NADPH was trapped by GSH, and this intermediate was believed to be involved in CYP2C9 inactivation. derived ortho-quinone 14-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 35583123-9 2022 A methysticin-derived ortho-quinone intermediate dependent on NADPH was trapped by GSH, and this intermediate was believed to be involved in CYP2C9 inactivation. NADP 62-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 35583123-9 2022 A methysticin-derived ortho-quinone intermediate dependent on NADPH was trapped by GSH, and this intermediate was believed to be involved in CYP2C9 inactivation. Glutathione 83-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 35583123-11 2022 Taken together, the present work demonstrated that methysticin was a mechanism-based inactivator of CYP2C9. methysticin 51-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 35629204-10 2022 These data might be relevant for implementation in the diverse clinical guidelines for the pharmacogenetic analysis of the CYP2C9 gene before treatment with different drugs, such as non-steroidal anti-inflammatory drugs, warfarin, phenytoin and statins. Warfarin 221-229 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 35629204-10 2022 These data might be relevant for implementation in the diverse clinical guidelines for the pharmacogenetic analysis of the CYP2C9 gene before treatment with different drugs, such as non-steroidal anti-inflammatory drugs, warfarin, phenytoin and statins. Phenytoin 231-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 35620290-14 2022 The dosage regimen, body mass index, CYP2C9 genetic polymorphism and fluconazole combination may had influences on siponimod pharmacokinetics. siponimod 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. 13cisra 28-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. Tretinoin 37-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 35545255-6 2022 In human hepatocytes (n=3), 13cisRA, atRA and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. 4-oxo-13cisra 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 35289081-7 2022 As a result, the marginal plasma exposure changes of typical CYP3A and CYP2C9 substrates were less than the bioequivalence threshold, indicating that anlotinib has a very low potential of causing clinically meaningful DDI through the inhibition of several major CYP enzymes. anlotinib 150-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 35246464-5 2022 The current study also demonstrated that selective inhibition sufficient for fm calculation was achieved by inhibitors of CYP1A2 (20 microM furafylline), CYP2C8 (40 microM montelukast), CYP2C9 (40 microM sulfaphenazole), CYP2C19 (3 microM (-)N-3-benzyl-phenobarbital) and CYP2D6 (5 microM quinidine). Sulfaphenazole 204-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 35246464-5 2022 The current study also demonstrated that selective inhibition sufficient for fm calculation was achieved by inhibitors of CYP1A2 (20 microM furafylline), CYP2C8 (40 microM montelukast), CYP2C9 (40 microM sulfaphenazole), CYP2C19 (3 microM (-)N-3-benzyl-phenobarbital) and CYP2D6 (5 microM quinidine). N-3-benzylphenobarbital 242-266 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 35246464-5 2022 The current study also demonstrated that selective inhibition sufficient for fm calculation was achieved by inhibitors of CYP1A2 (20 microM furafylline), CYP2C8 (40 microM montelukast), CYP2C9 (40 microM sulfaphenazole), CYP2C19 (3 microM (-)N-3-benzyl-phenobarbital) and CYP2D6 (5 microM quinidine). Quinidine 289-298 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 35426132-0 2022 The Impact of CYP2C9*11 Genetic Polymorphism on the Pharmacokinetics of Phenytoin and (S)-Warfarin. Phenytoin 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 35138605-11 2022 After combined treatment of HepG2 cells with warfarin + PNS, CYP1A2 expression was upregulated (P < 0.05) and CYP3A4 was downregulated (P < 0.05) but there was no effect on CYP2C9. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 175-181 35439929-1 2022 OBJECTIVE: To explore the role of genetic testing of VKORC1 and CYP2C9 in determining the dosage of warfarin after aortic valve replacement. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 35247543-9 2022 CONCLUSIONS: Observed inhibition of metabolic enzymes indicated that remibrutinib is a weak inhibitor of CYP3A4 and CYP2C9 and is not a clinically relevant inhibitor of uptake and efflux transporters, except for intestinal P-glycoprotein and breast cancer resistance protein inhibition. Remibrutinib 69-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 35380455-4 2022 The most studied gene-drug pairs were clopidogrel and warfarin associated with cytochrome p450 and vitamin K epoxide reductase complex subunit 1 genes (CYP2C19, CYP2C9 and VKORC1), classified as critically low quality. Clopidogrel 38-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 35380455-4 2022 The most studied gene-drug pairs were clopidogrel and warfarin associated with cytochrome p450 and vitamin K epoxide reductase complex subunit 1 genes (CYP2C19, CYP2C9 and VKORC1), classified as critically low quality. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 35426132-0 2022 The Impact of CYP2C9*11 Genetic Polymorphism on the Pharmacokinetics of Phenytoin and (S)-Warfarin. Warfarin 86-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 35426132-5 2022 The objective of the current study was to characterize the in-vivo activity of CYP2C9 among carriers of CYP2C9*11, one of the "African" alleles and the fourth most common CYP2C9 variant allele among Caucasians by using two prototype substrates, phenytoin and (S)-warfarin. Phenytoin 245-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 35426132-5 2022 The objective of the current study was to characterize the in-vivo activity of CYP2C9 among carriers of CYP2C9*11, one of the "African" alleles and the fourth most common CYP2C9 variant allele among Caucasians by using two prototype substrates, phenytoin and (S)-warfarin. Warfarin 259-271 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 35426132-11 2022 Among patients of African ancestry CYP2C9*11 genetic analysis should be considered prior to prescribing of narrow therapeutic window drugs such as phenytoin, warfarin, NSAIDs or siponimod. Phenytoin 147-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 35426132-11 2022 Among patients of African ancestry CYP2C9*11 genetic analysis should be considered prior to prescribing of narrow therapeutic window drugs such as phenytoin, warfarin, NSAIDs or siponimod. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 35426132-11 2022 Among patients of African ancestry CYP2C9*11 genetic analysis should be considered prior to prescribing of narrow therapeutic window drugs such as phenytoin, warfarin, NSAIDs or siponimod. siponimod 178-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 35456605-0 2022 Population Pharmacokinetic (Pop-PK) Analysis of Torsemide in Healthy Korean Males Considering CYP2C9 and OATP1B1 Genetic Polymorphisms. Torsemide 48-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 35365981-0 2022 Association of VKORC1 and CYP2C9 single-nucleotide polymorphisms with warfarin dose adjustment in Saudi patients. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 35365981-2 2022 Previous reports have suggested that polymorphisms in VKORC1 and CYP2C9 genes could influence warfarin therapy. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 35365981-8 2022 Similarly, there was a significant reduction in warfarin daily maintenance dose among CYP2C9*1/*3 and CYP2C9*1/*2 groups compared to CYP2C9*1/*1. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 35365981-8 2022 Similarly, there was a significant reduction in warfarin daily maintenance dose among CYP2C9*1/*3 and CYP2C9*1/*2 groups compared to CYP2C9*1/*1. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 35365981-10 2022 CONCLUSIONS: Similar to other populations, the VKORC1 and CYP2C9 gene polymorphisms are significantly associated with warfarin dosage in Saudi patients. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 35365981-11 2022 The presence of at least one copy of the mutant alleles for VKORC1 -1173C>T, CYP2C9*2, and CYP2C9*3 is associated with a significant reduction in warfarin maintenance dose. Warfarin 146-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 35365981-11 2022 The presence of at least one copy of the mutant alleles for VKORC1 -1173C>T, CYP2C9*2, and CYP2C9*3 is associated with a significant reduction in warfarin maintenance dose. Warfarin 146-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 35108398-0 2022 Warfarin Dosing in Patients with CYP2C9*5 Variant Alleles. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 35108398-2 2022 The cytochrome P450 2C9*5 (CYP2C9*5) allele is found almost exclusively in populations of African ancestry, and in-vitro studies suggest CYP2C9*5 is associated with reduced clearance of warfarin. Warfarin 186-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 35108398-2 2022 The cytochrome P450 2C9*5 (CYP2C9*5) allele is found almost exclusively in populations of African ancestry, and in-vitro studies suggest CYP2C9*5 is associated with reduced clearance of warfarin. Warfarin 186-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 35108398-4 2022 In this multi-centered study of 2298 patients (49% female, 35% Black) taking warfarin, we quantified the association between the CYP2C9*5 allele and warfarin requirements. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 35108398-6 2022 Without taking CYP2C9*5 into account, pharmacogenetic algorithms that include other SNPs overestimated the warfarin dose by 30% (95% CI (19%-40%), p<0.001), an average of 1.87 mg/d (SD 1.64) in heterozygotes (p < 0.001). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 35108398-9 2022 Patients carrying CYP2C9*5 require a clinically relevant reduction in warfarin dose. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 35115300-8 2022 Oral (130 mg) or inhaled (75 mg) THC was predicted to precipitate interactions with drugs predominately metabolized by CYP2C9 (diclofenac, 6.6 or 2.3, respectively) >3A (midazolam, 1.8) >1A2 (theophylline, 1.4). Dronabinol 33-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 35115300-8 2022 Oral (130 mg) or inhaled (75 mg) THC was predicted to precipitate interactions with drugs predominately metabolized by CYP2C9 (diclofenac, 6.6 or 2.3, respectively) >3A (midazolam, 1.8) >1A2 (theophylline, 1.4). Diclofenac 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 35115300-8 2022 Oral (130 mg) or inhaled (75 mg) THC was predicted to precipitate interactions with drugs predominately metabolized by CYP2C9 (diclofenac, 6.6 or 2.3, respectively) >3A (midazolam, 1.8) >1A2 (theophylline, 1.4). Theophylline 192-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 35456605-9 2022 According to the genetic polymorphisms of OATP1B1 and CYP2C9, significant associations were found in the V/F, CL/F, and CL2/F of torsemide. Torsemide 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 35456605-11 2022 Considering that torsemide is a substrate for CYP2C9 and OATP1B1, it was important to search for genetic polymorphisms in CYP2C9 and OATP1B1 as covariates to explain the PK diversity of torsemide between individuals. Torsemide 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 35456605-11 2022 Considering that torsemide is a substrate for CYP2C9 and OATP1B1, it was important to search for genetic polymorphisms in CYP2C9 and OATP1B1 as covariates to explain the PK diversity of torsemide between individuals. Torsemide 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 35456605-14 2022 These results suggested that the phenotypes of CYP2C9 and OATP1B1 produced significant differences in torsemide PKs. Torsemide 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 35456605-15 2022 Considering that CYP2C9 and OATP1B1 phenotypes as covariates affected different PK parameters of torsemide, it could be inferred that torsemide"s cell membrane permeation process by OATP1B1 and the metabolic process by CYP2C9 could independently affect each other in vivo without interplay. Torsemide 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 35456605-15 2022 Considering that CYP2C9 and OATP1B1 phenotypes as covariates affected different PK parameters of torsemide, it could be inferred that torsemide"s cell membrane permeation process by OATP1B1 and the metabolic process by CYP2C9 could independently affect each other in vivo without interplay. Torsemide 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 35456605-15 2022 Considering that CYP2C9 and OATP1B1 phenotypes as covariates affected different PK parameters of torsemide, it could be inferred that torsemide"s cell membrane permeation process by OATP1B1 and the metabolic process by CYP2C9 could independently affect each other in vivo without interplay. Torsemide 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 35456605-15 2022 Considering that CYP2C9 and OATP1B1 phenotypes as covariates affected different PK parameters of torsemide, it could be inferred that torsemide"s cell membrane permeation process by OATP1B1 and the metabolic process by CYP2C9 could independently affect each other in vivo without interplay. Torsemide 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 35401180-6 2022 Compared with the control group, the expression of P-glycoprotein (P-gp), CYP1A2, CYP2C9, CYP3A4, and nuclear receptor, PXR, were altered in the amoxicillin -treated groups. Amoxicillin 145-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 35345577-0 2022 Association of AGTR1 A1166C and CYP2C9*3 Gene Polymorphisms with the Antihypertensive Effect of Valsartan. Valsartan 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 35345577-2 2022 The present study aimed to investigate whether the antihypertensive effect of the ARB drug valsartan was associated with angiotensin II type 1 receptor (AGTR1) gene polymorphism (A1166 C) and cytochrome P450 enzyme 2C9 (CYP2C9) gene polymorphism (CYP2C9*3). Valsartan 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-218 35345577-2 2022 The present study aimed to investigate whether the antihypertensive effect of the ARB drug valsartan was associated with angiotensin II type 1 receptor (AGTR1) gene polymorphism (A1166 C) and cytochrome P450 enzyme 2C9 (CYP2C9) gene polymorphism (CYP2C9*3). Valsartan 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 247-253 35345577-2 2022 The present study aimed to investigate whether the antihypertensive effect of the ARB drug valsartan was associated with angiotensin II type 1 receptor (AGTR1) gene polymorphism (A1166 C) and cytochrome P450 enzyme 2C9 (CYP2C9) gene polymorphism (CYP2C9*3). Valsartan 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Phenytoin 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 35293300-9 2022 ZY12201 Ki values were 0.38, 0.25, 0.07, 0.01, 0.06, 0.02, 7.13, 0.03 and 0.003 muM for CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4/5 (substrate: testosterone) and CYP3A4/5 (substrate: midazolam), respectively. zy12201 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 35293300-10 2022 Time-dependant CYP inhibition potential of ZY12201 was assessed against CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 and no apparent IC50 shift was observed. zy12201 43-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 35293300-14 2022 ZY12201 is a non-time-dependent inhibitor of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4/5. zy12201 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 35202038-0 2022 Association between CYP2C9, VKORC1, VDR, and APOE genotypes on warfarin maintenance and response during initial anticoagulation for Chinese patients with heart valve replacement. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 35335853-4 2022 The Michaelis-Menten kinetics of voriconazole N-oxide (NO) formation, the major circulating metabolite, by CYP2C19, CYP2C9 and CYP3A4, was determined in incubations of human recombinant CYP enzymes and liver and intestine microsomes. Voriconazole N-Oxide 33-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 35062050-13 2022 In the NAFLD model, Omeprazole maintained an expected induction of CYP1A1, however Phenytoin and Rifampicin showed elevated induction of CYP2B6 and CYP2C9 compared to healthy cultures. Rifampin 97-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 35342886-8 2022 CYP2C9-reduced metabolism genotypes were associated with losartan response and uncontrolled hypertension (odds ratio (OR), 5.2; 95% CI, 1.9 to 14.7). Losartan 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 35078445-12 2022 In vitro, we further confirmed that curcumin significantly downregulated the expression of AURKA, CDK1, and TOP2A genes, while significantly upregulated the expression of CYP2B6, CYP2C9, and CYP3A4 genes. Curcumin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 35078445-13 2022 CONCLUSIONS: Our results provided a novel panel of AURKA, CDK1, TOP2A, CYP2C9, and CYP3A4 candidate genes for curcumin related chemotherapy of hepatocellular carcinoma. Curcumin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 35000550-3 2021 In vitro, CC-90001 glucuronidation was catalyzed by UGT1A9, UGT1A4, and UGT1A1, while oxidative metabolism was primarily mediated by CYP3A4/5 with minor contributions from CYP1A2, CYP2C9, CYP2B6, and CYP2D6.2. CC-90001 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 180-186 35282004-10 2022 Twenty-one cytochrome P450 (CYP2C9, CYP2C18, and CYP2C19) "metabolism" polymorphisms in locus 10q23.33 were significantly associated with higher beta-carotene concentration. beta Carotene 145-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 35282004-12 2022 Zeaxanthin, lycopene, and beta-carotene serum concentrations might depend on genetic variation in NR1H3, APOB, RDH12 and CYP2C9, CYP2C18, and CYP2C19 genes. Zeaxanthins 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 35282004-12 2022 Zeaxanthin, lycopene, and beta-carotene serum concentrations might depend on genetic variation in NR1H3, APOB, RDH12 and CYP2C9, CYP2C18, and CYP2C19 genes. beta Carotene 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 35000550-4 2021 CC-90001 in vitro inhibits CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 <= 55% at 100 muM, and the inhibition was negligible at <=30 muM. CC-90001 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 2827463-0 1988 Human cytochrome P-450 PB-1: a multigene family involved in mephenytoin and steroid oxidations that maps to chromosome 10. Mephenytoin 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-27 35169023-1 2022 BACKGROUND: The aim of this study was to investigate the effect of CYB2B6 (c.516G>T, rs3745274), CYP2C9 (c.1075A>C, rs1057910) and UGT1A9 (c.98T>C, rs72551330) polymorphisms on the pharmacokinetics of single-drug propofol in adult patients undergoing intravenous sedation. Propofol 213-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 2827463-0 1988 Human cytochrome P-450 PB-1: a multigene family involved in mephenytoin and steroid oxidations that maps to chromosome 10. Steroids 76-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-27 33346393-0 2021 A network meta-analysis of CYP2C9, CYP2C9 with VKORC1 and CYP2C9 with VKORC1 and CYP4F2 genotype-based warfarin dosing strategies compared to traditional. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 33336382-7 2021 In addition, metamizole weakly induced CYP2C9 (decrease in the flurbiprofen AUC by 22%) and moderately CYP2C19 (decrease in the omeprazole AUC by 66%) but did not alter CYP2D6 activity. Dipyrone 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 33336382-7 2021 In addition, metamizole weakly induced CYP2C9 (decrease in the flurbiprofen AUC by 22%) and moderately CYP2C19 (decrease in the omeprazole AUC by 66%) but did not alter CYP2D6 activity. Flurbiprofen 63-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 33346393-0 2021 A network meta-analysis of CYP2C9, CYP2C9 with VKORC1 and CYP2C9 with VKORC1 and CYP4F2 genotype-based warfarin dosing strategies compared to traditional. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 33346393-7 2021 CYP2C9-based warfarin dosing was associated with a shorter time to first therapeutic INR (WMD: -2.73, 95% CI: -3.41, -2.05) and stable INR/warfarin dose (WMD: -8.1, 95% CI: -12.54, -3.66). Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33346393-7 2021 CYP2C9-based warfarin dosing was associated with a shorter time to first therapeutic INR (WMD: -2.73, 95% CI: -3.41, -2.05) and stable INR/warfarin dose (WMD: -8.1, 95% CI: -12.54, -3.66). Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33346393-8 2021 CYP2C9 and VKORC1 were observed with a shorter time to first therapeutic INR (WMD: -1.92, 95% CI: -3.23, -0.61) and stable INR/warfarin dose (WMD: -4.6, 95% CI: -6.87, -2.34) along with a longer TTR (%) (WMD: 3.91, 95% CI: 1.18, 6.63). Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33346393-11 2021 WHAT IS NEW AND CONCLUSION: The present evidence is supportive of personalizing warfarin dose based only on CYP2C9 and VKORC1 genotypes compared to traditional strategies. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 33961833-3 2021 Our data indicated that miconazole potently inhibited phenytoin hydroxylation in both pooled HLMs and recombinant cytochrome P450 2C9 (CYP2C9) with the Ki values of 125 +-7 nM and 30 +- 2 nM, respectively. Miconazole 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-133 33955686-0 2021 Effect of GLPG1205, a GPR84 Modulator, on CYP2C9, CYP2C19, and CYP1A2 Enzymes: In Vitro and Phase 1 Studies. GLPG1205 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. carfentanil 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. Norcarfentanil 125-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. Piperidines 185-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. 2-phenylethyl acetate 219-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. Ketones 239-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33652071-8 2021 These findings indicate that CYP3A5, CYP3A4, CYP2C8 and CYP2C9 play a major role in the transformation of carfentanil to M1 (norcarfentanil), M11, M13 and M15 through N-dealkylation of piperidine ring, hydroxylation of phenethyl group and ketone formation on phenethyl linker by human liver micrsomes. 2-phenylethyl acetate 259-268 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33955686-3 2021 In vitro experiments indicated weak inhibition of CYP1A2 and moderate but reversible inhibition of CYP2C9 and CYP2C19 by GLPG1205. GLPG1205 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 33961833-3 2021 Our data indicated that miconazole potently inhibited phenytoin hydroxylation in both pooled HLMs and recombinant cytochrome P450 2C9 (CYP2C9) with the Ki values of 125 +-7 nM and 30 +- 2 nM, respectively. Miconazole 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 33961833-3 2021 Our data indicated that miconazole potently inhibited phenytoin hydroxylation in both pooled HLMs and recombinant cytochrome P450 2C9 (CYP2C9) with the Ki values of 125 +-7 nM and 30 +- 2 nM, respectively. Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-133 33961833-3 2021 Our data indicated that miconazole potently inhibited phenytoin hydroxylation in both pooled HLMs and recombinant cytochrome P450 2C9 (CYP2C9) with the Ki values of 125 +-7 nM and 30 +- 2 nM, respectively. Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 33632714-3 2021 M1L is present at a minor allele frequency of 6.3% in Yup"ik Alaska Native people and, thus, can contribute to the risk of an adverse drug response from narrow therapeutic index CYP2C9 substrates, such as (S)-warfarin. Warfarin 205-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 33958549-6 2021 Our results showed that 13 SNPs on 6 genes (CYP2C9, VKORC1, ABCB1, PROC, EPHX1 and SETD1A) were associated with the individual stable warfarin dose. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 33958549-9 2021 We discovered that, genetic variants in CYP2C9, VKORC1, ABCB1, PROC, EPHX1 and SETD1A may affect the stable warfarin dose requirement in Han Chinese patients with valvular disease. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 33632714-9 2021 Here, we validated (S)-naproxen as a CYP2C9 probe substrate to characterize the in vivo functional activity of the CYP2C9 Leu1 variant. Naproxen 19-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 33632714-5 2021 We first confirmed the selectivity of (S)-naproxen O-demethylation by CYP2C9 using activity phenotyped human liver microsomes and selective CYP inhibitors, then developed and validated a novel LC/MS method for simultaneous quantification of (S)-naproxen, (S)-O-desmethyl naproxen, and naproxen acyl glucuronide in human urine. Naproxen 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 33632714-9 2021 Here, we validated (S)-naproxen as a CYP2C9 probe substrate to characterize the in vivo functional activity of the CYP2C9 Leu1 variant. Naproxen 19-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 33610566-8 2021 Moreover, isobavachalcone demonstrated broad-spectrum inhibitory effects against CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, UGT1A1, UGT1A9, UGT2B7 with IC50 values of 1.08-9.78 muM. isobavachalcone 10-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 33509019-0 2021 Effect of CYP2C9 genetic polymorphism and breviscapine on losartan pharmacokinetics in healthy subjects. Losartan 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 33509019-7 2021 Individuals with genotype CYP2C9*1/*3 showed a significant increase in AUC (0-36) (2335 +- 851.8 ng h ml-1 vs. 1927 +- 949.5 ng h ml-1, p < 0.05) and AUC (0- ) (2363 +- 875.6 ng h ml-1 vs. 1966 +- 966.1 ng h ml-1, p < 0.05) of E-3174 after breviscapine treatment compared to the placebo group. breviscapine 240-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 33509019-9 2021 The activity of CYP2C9 enzyme to losartan metabolism was more significant in subjects with CYP2C9*1/*3 than those with CYP2C9*1/*1 genotype. Losartan 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 33509019-9 2021 The activity of CYP2C9 enzyme to losartan metabolism was more significant in subjects with CYP2C9*1/*3 than those with CYP2C9*1/*1 genotype. Losartan 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 33509019-9 2021 The activity of CYP2C9 enzyme to losartan metabolism was more significant in subjects with CYP2C9*1/*3 than those with CYP2C9*1/*1 genotype. Losartan 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 33840516-0 2021 Association Between the CYP2C9 Genotype and Hypoglycemia Among Patients With Type 2 Diabetes Receiving Sulfonylurea Treatment: A Meta-analysis. Sulfonylurea Compounds 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 33663326-2 2021 To determine the drug-drug interaction risk for cytochrome P450, and SLC and ABC transporters, brexpiprazole and its metabolite, DM-3411 were assessed in this in vitro investigation.Brexpiprazole exhibited weak inhibitory effects (IC50 >13 mumol/L) on CYP2C9, CYP2C19, CYP2D6 and CYP3A4 activities, but had moderate inhibitor activity on CYP2B6 (IC50 8.19 mumol/L). brexpiprazole 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 252-258 33890704-6 2021 Treatment with metamizole, a CAR-dependent inducer of CYP2B6, CYP2C9, CYP2C19 and CYP3A4, accentuated the effect of deglucuronidation on AUC and MR. Dipyrone 15-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 33859491-0 2021 CYP2C9*3/*3 Gene Expression Affects the Total and Free Concentrations of Valproic Acid in Pediatric Patients with Epilepsy. Valproic Acid 73-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33859491-7 2021 VPA"s daily doses and free drug concentrations were significantly lower in the CYP2C9 *3/*3 genotype group versus the CYP2C9 *1/*3 and CYP2C9 *1/*1 groups (P<0.05). Valproic Acid 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 33859491-7 2021 VPA"s daily doses and free drug concentrations were significantly lower in the CYP2C9 *3/*3 genotype group versus the CYP2C9 *1/*3 and CYP2C9 *1/*1 groups (P<0.05). Valproic Acid 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 33859491-7 2021 VPA"s daily doses and free drug concentrations were significantly lower in the CYP2C9 *3/*3 genotype group versus the CYP2C9 *1/*3 and CYP2C9 *1/*1 groups (P<0.05). Valproic Acid 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 33859491-11 2021 Additionally, CYP2C9 *3/*3 expression affects VPA metabolism. Valproic Acid 46-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 33840516-2 2021 Because impaired catalytic function is likely to affect sulfonylurea metabolism, it is predictable that CYP2C9 loss-of-function alleles may increase the risk of sulfonylurea-induced hypoglycemia. Sulfonylurea Compounds 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 33840516-2 2021 Because impaired catalytic function is likely to affect sulfonylurea metabolism, it is predictable that CYP2C9 loss-of-function alleles may increase the risk of sulfonylurea-induced hypoglycemia. Sulfonylurea Compounds 161-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 33840516-3 2021 This systematic review and meta-analysis aimed to assess the association between CYP2C9 genotype and hypoglycemia among patients with type 2 diabetes mellitus (T2DM) receiving sulfonylurea. Sulfonylurea Compounds 176-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 33840516-4 2021 METHODS: We searched for studies on the association between CYP2C9 genotype and sulfonylurea-induced hypoglycemia among patients with T2DM, published through August 7, 2020, using PubMed, Web of Science, and EMBASE. Sulfonylurea Compounds 80-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 33840516-7 2021 The CYP2C9 variant alleles were associated with an increase in sulfonylurea-induced hypoglycemia compared with wild-type homozygote (OR = 1.24; 95% CI, 1.03-1.48). Sulfonylurea Compounds 63-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 33840516-9 2021 IMPLICATIONS: On the basis of these results, CYP2C9 genotyping may be useful for predicting the risk of hypoglycemia during sulfonylurea treatment for T2DM. Sulfonylurea Compounds 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 28520374-0 2012 Phenytoin Therapy and HLA-B*15:02 and CYP2C9 Genotypes Phenytoin is an antiseizure medication used for the prevention of focal seizures and generalized tonic-clonic convulsions (1). Phenytoin 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 28520374-4 2012 CYP2C9 is one of the main enzymes involved in the metabolism of phenytoin, and variant CYP2C9 alleles are known to influence phenytoin drug levels. Phenytoin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28520374-4 2012 CYP2C9 is one of the main enzymes involved in the metabolism of phenytoin, and variant CYP2C9 alleles are known to influence phenytoin drug levels. Phenytoin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 28520374-4 2012 CYP2C9 is one of the main enzymes involved in the metabolism of phenytoin, and variant CYP2C9 alleles are known to influence phenytoin drug levels. Phenytoin 125-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28520374-4 2012 CYP2C9 is one of the main enzymes involved in the metabolism of phenytoin, and variant CYP2C9 alleles are known to influence phenytoin drug levels. Phenytoin 125-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 28520374-12 2012 The label also mentions that variant CYP2C9 alleles may contribute to unusually high levels of phenytoin (1). Phenytoin 95-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 33421896-3 2021 The redox properties of the immobilised proteins were investigated by cyclic voltammetry and electrocatalysis was measured in presence of the largely used anticoagulant drug S-warfarin, marker substrate for CYP2C9. Warfarin 174-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 207-213 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. didodecyldimethylammonium 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. didodecyldimethylammonium 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. didodecyldimethylammonium 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Carbon 88-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Carbon 88-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Carbon 88-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Oxygen 147-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Oxygen 147-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33450066-5 2021 KEY RESULTS: Using microsomes containing recombinant CYPs expressed in human vascular tissues, we show that nitrite is released from NTG and PETN with CYP2C9 and CYP2C8 being most efficient. Nitrites 108-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 33421896-4 2021 Immobilisation of the CYP2C9-FLD, CYP2C9*2-FLD and CYP2C9*3-FLD on DDAB modified glassy carbon electrodes showed well defined redox couples on the oxygen-free cyclic voltammograms and mid-point potentials of all enzymes were calculated. Oxygen 147-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 33675325-7 2021 For all patients, pharmacogenetic diagnostic for the genes CYP2C9, CYP2C19 and NAT2, which are involved in metamizole metabolism and degradation of toxic metabolites, was initiated. Dipyrone 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 33394101-2 2021 The study investigated the drug-drug interaction (DDI) potential of ceritinib when coadministered with midazolam and warfarin as probe substrates for CYP3A and CYP2C9 activity, respectively. ceritinib 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 33394101-11 2021 CONCLUSION: Ceritinib is a strong CYP3A inhibitor and a weak CYP2C9 inhibitor. ceritinib 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 33849700-0 2021 The inhibition of CYP1A2, CYP2C9, and CYP2D6 by pterostilbene in human liver microsomes. pterostilbene 48-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 33849700-1 2021 This study used human liver microsomes to assess pterostilbene"s effect on the metabolic activity of cytochrome P450 (CYP) 1A2, CYP2C9, and CYP2D6. pterostilbene 49-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 33849700-4 2021 In this study, pterostilbene inhibited CYP1A2, CYP2C9, and CYP2D6"s metabolic activities in vitro. pterostilbene 15-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 33849700-5 2021 CYP2C9"s activity was most significantly inhibited by pterostilbene; its IC50 value was 0.12+-0.04 muM. pterostilbene 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33849700-7 2021 The finding that suggests that pterostilbene has the potential to interact with CYP2C9 substrates in vivo. pterostilbene 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 33731883-4 2021 Genetic polymorphisms of CYP2C9, the main enzyme involved in the metabolism of sulfonylureas, have been associated with the risk of severe hypoglycaemia, particularly in poor metabolizers carrying CYP2C9 *3/*3 genotype, and especially in the case of patients treated with glimepiride. Sulfonylurea Compounds 79-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 33731883-4 2021 Genetic polymorphisms of CYP2C9, the main enzyme involved in the metabolism of sulfonylureas, have been associated with the risk of severe hypoglycaemia, particularly in poor metabolizers carrying CYP2C9 *3/*3 genotype, and especially in the case of patients treated with glimepiride. Sulfonylurea Compounds 79-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 33731883-4 2021 Genetic polymorphisms of CYP2C9, the main enzyme involved in the metabolism of sulfonylureas, have been associated with the risk of severe hypoglycaemia, particularly in poor metabolizers carrying CYP2C9 *3/*3 genotype, and especially in the case of patients treated with glimepiride. glimepiride 272-283 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 33731883-4 2021 Genetic polymorphisms of CYP2C9, the main enzyme involved in the metabolism of sulfonylureas, have been associated with the risk of severe hypoglycaemia, particularly in poor metabolizers carrying CYP2C9 *3/*3 genotype, and especially in the case of patients treated with glimepiride. glimepiride 272-283 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 33731883-5 2021 The objectives of the present study were to evaluate the potential clinical and economic outcomes of using CYP2C9 genotype data to guide the management of SU regimen in patients initiating glimepiride therapy, and to identify factors affecting the cost-effectiveness of this treatment scheme. glimepiride 189-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 33731883-9 2021 In conclusion, the potential cost of CYP2C9 genotype-guided dosing for glimepiride therapy is relatively high, and associated with modest improvements with respect to the number of hypoglycaemia avoided, as compared with standard dosing. glimepiride 71-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 33243500-3 2021 The results indicated that Mo or/and Cd induced CYP1A1, CYP1B1, CYP2C9, CYP3A8 and CYP4B1 mRNA levels, decreased superoxide dismutase (SOD), catalase (CAT) activities and glutathione peroxidase (GSH-Px) content, and increased malondialdehyde (MDA) and hydrogen peroxide (H2O2) contents. Cadmium 37-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 33556367-8 2021 In contrast, the binding of both SOR and SNO to active site residues in the closely related human CYP2C9 enzyme was similar, as were the IC50s determined against CYP2C9-mediated losartan oxidation. Sorafenib 33-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 33556367-8 2021 In contrast, the binding of both SOR and SNO to active site residues in the closely related human CYP2C9 enzyme was similar, as were the IC50s determined against CYP2C9-mediated losartan oxidation. Losartan 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 33754183-0 2021 S-warfarin limited sampling strategy with a population pharmacokinetic approach to estimate exposure and cytochrome P450 (CYP) 2C9 activity in healthy adults. Warfarin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-130 33754183-1 2021 PURPOSE: S-warfarin is used to phenotype cytochrome P450 (CYP) 2C9 activity. Warfarin 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-66 33754183-11 2021 During CYP2C9 induction, S-warfarin LSMs had unacceptable %MPE, %MAE, and %RMSE. Warfarin 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 7-13 33754183-12 2021 CONCLUSIONS: Phenotyping studies with S-warfarin in healthy subjects can utilize a single- and/or a 2-timepoint LSM with a population PK approach to estimate constitutive CYP2C9 activity. Warfarin 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 33740842-2 2021 Cannabinoids are subjected to a significant first-pass metabolism in the liver involving the cytochrome P450 enzymes (CYP) 2C9, CYP2C19 and CYP3A4 [2];. Cannabinoids 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-126 33727862-1 2021 Purpose: To assess the association between VKORC1 and CYP2C9 variants and the incidence of adverse drug reactions in warfarin-treated patients in a real-world setting. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 33675325-14 2021 In conclusion, metamizole-induced agranulocytosis most likely was a consequence of the underlying genetical predisposition, that is, polymorphisms in the genes NAT2, CYP2C9 and CYP2C19. Dipyrone 15-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 32736411-1 2021 AIMS: We evaluated the potential effect of sonidegib at an oral dose of 800 mg once daily (QD) on the pharmacokinetics (PK) of the probe drugs warfarin (CYP2C9) and bupropion (CYP2B6). sonidegib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 33191909-1 2021 OBJECTIVE: We attempted to examine the applicability of a population pharmacokinetic-pharmacodynamic (PK-PD) model describing the metabolic interaction between warfarin and sorafenib due to CYP2C9 inhibition and to predict the plasma concentrations of sorafenib and S-warfarin, the international normalized ratio (INR), and the optimal maintenance dose of warfarin in the presence of sorafenib in vivo. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 33278335-1 2021 VKORC1 and CYP2C9 genotypes explain less variability in warfarin dose requirements in African Americans (AAs) compared to Europeans. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 33278335-2 2021 Variants in BCKDK and GATA-4 gene regions, purported to regulate VKORC1 and CYP2C9 expression, have been shown to play an important role in warfarin dose requirements in Europeans and Asians, respectively. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 33191909-1 2021 OBJECTIVE: We attempted to examine the applicability of a population pharmacokinetic-pharmacodynamic (PK-PD) model describing the metabolic interaction between warfarin and sorafenib due to CYP2C9 inhibition and to predict the plasma concentrations of sorafenib and S-warfarin, the international normalized ratio (INR), and the optimal maintenance dose of warfarin in the presence of sorafenib in vivo. Sorafenib 173-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 32918831-2 2021 In vitro data showed pexidartinib"s potential to inhibit and induce cytochrome P450 (CYP) 3A, inhibit CYP2C9, CYP2C19 and P-glycoprotein (P-gp). pexidartinib 21-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 32918831-3 2021 Herein, 2 open-label, single-sequence, crossover studies evaluated the drug-drug interaction potential of pexidartinib on CYP enzymes (CYP2C9, CYP2C19, and CYP3A) and P-gp. pexidartinib 106-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 32918831-11 2021 These results indicate that pexidartinib is a moderate inducer of CYP3A and a weak inhibitor of CYP2C9 and does not significantly affect CYP2C19-mediated metabolism or P-gp transport. pexidartinib 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 33253783-5 2021 Scaling of relative P450 efficiencies based on average liver concentration revealed that CYP2C8 dominates the sudoxicam bioactivation pathway and CYP2C9 dominates meloxicam detoxification. Meloxicam 163-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 33600055-4 2021 In this study, we systematically evaluated the effects of protein crystal structural models, scoring functions, heme forms, conserved active site water molecule, and protein flexibility on SOMs prediction of CYP2C9 substrates. Heme 112-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 33600055-4 2021 In this study, we systematically evaluated the effects of protein crystal structural models, scoring functions, heme forms, conserved active site water molecule, and protein flexibility on SOMs prediction of CYP2C9 substrates. Water 146-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 208-214 33341662-0 2021 Effects of acid and lactone forms of statins on S-warfarin 7-hydroxylation catalyzed by human liver microsomes and recombinant CYP2C9 variants (CYP2C9.1 and CYP2C9.3). Warfarin 48-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 33747646-4 2021 These medications inhibit two vital enzymes of cytochrome P-450, CYP2C9 and CYP3A4, potentiating the effects of ATRA on calcium metabolism. Tretinoin 112-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 33747646-4 2021 These medications inhibit two vital enzymes of cytochrome P-450, CYP2C9 and CYP3A4, potentiating the effects of ATRA on calcium metabolism. Calcium 120-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 33540768-1 2021 Cytochrome P450 2C9 (CYP2C9) is an important drug-metabolizing enzyme that contributes to the metabolism of approximately 15% of clinically used drugs, including warfarin, which is known for its narrow therapeutic window. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 33540768-1 2021 Cytochrome P450 2C9 (CYP2C9) is an important drug-metabolizing enzyme that contributes to the metabolism of approximately 15% of clinically used drugs, including warfarin, which is known for its narrow therapeutic window. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 33540768-4 2021 These CYP2C9 variants were heterologously expressed in 293FT cells, and the kinetic parameters (Km, kcat, Vmax, catalytic efficiency, and CLint) of (S)-warfarin 7-hydroxylation and tolbutamide 4-hydroxylation were estimated. Warfarin 148-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 33540768-6 2021 A strong correlation was found in catalytic efficiencies between (S)-warfarin 7-hydroxylation and tolbutamide 4-hydroxylation among all studied CYP2C9 variants. Warfarin 65-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 33540768-6 2021 A strong correlation was found in catalytic efficiencies between (S)-warfarin 7-hydroxylation and tolbutamide 4-hydroxylation among all studied CYP2C9 variants. Tolbutamide 98-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 33540768-8 2021 Our findings could clarify a part of discrepancies among genotype-phenotype associations based on the novel CYP2C9 rare allelic variants and could, therefore, improve personalized medicine, including the selection of the appropriate warfarin dose. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 33341662-1 2021 The inhibition of CYP2C9-mediated warfarin metabolism by acid or lactone forms of statin converted in the body and effects of CYP2C9 genetic variants on their inhibition are not fully understood. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 33341662-1 2021 The inhibition of CYP2C9-mediated warfarin metabolism by acid or lactone forms of statin converted in the body and effects of CYP2C9 genetic variants on their inhibition are not fully understood. Lactones 65-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 33341662-7 2021 These results indicated that lactone forms of statins other than fluvastatin showed more potent inhibition of CYP2C9-catalyzed S-warfarin 7-hydroxylation than the corresponding acid forms. Lactones 29-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 33341662-7 2021 These results indicated that lactone forms of statins other than fluvastatin showed more potent inhibition of CYP2C9-catalyzed S-warfarin 7-hydroxylation than the corresponding acid forms. Fluvastatin 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 33341662-7 2021 These results indicated that lactone forms of statins other than fluvastatin showed more potent inhibition of CYP2C9-catalyzed S-warfarin 7-hydroxylation than the corresponding acid forms. Warfarin 127-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 33341662-8 2021 Furthermore, our results indicated that Ile359Leu substitution in CYP2C9 affected the inhibitory potencies of statins. ile359leu 40-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 33259822-5 2021 The formation of MFA-quinoneimine by recombinant CYP1A2 and CYP2C9 was significantly decreased by the addition of human liver cytosol, and the extent of the decrease in the metabolite formed by CYP1A2 was larger than that by CYP2C9. mfa-quinoneimine 17-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 32779747-1 2021 Phenytoin is an antiepileptic drug with a narrow therapeutic index and large inter-patient pharmacokinetic variability, partly due to genetic variation in CYP2C9. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 32779747-3 2021 We summarize evidence from the published literature supporting these associations and provide therapeutic recommendations for the use of phenytoin based on CYP2C9 and/or HLA-B genotypes (updates on cpicpgx.org). Phenytoin 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 32869328-4 2021 A clinical study in healthy subjects (n = 27) evaluated the inhibition of CYP3A4, CYP2C8, and CYP2C9 in vivo by administering single doses of probe CYP substrates (midazolam, pioglitazone, and tolbutamide) alone and in combination with relacorilant (350 mg). Midazolam 164-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 32869328-4 2021 A clinical study in healthy subjects (n = 27) evaluated the inhibition of CYP3A4, CYP2C8, and CYP2C9 in vivo by administering single doses of probe CYP substrates (midazolam, pioglitazone, and tolbutamide) alone and in combination with relacorilant (350 mg). Tolbutamide 193-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 33039464-11 2021 LoT positively predicts genomic predisposition (CYP2C9*2) for Bosentan-induced cholestasis. Bosentan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 33259822-5 2021 The formation of MFA-quinoneimine by recombinant CYP1A2 and CYP2C9 was significantly decreased by the addition of human liver cytosol, and the extent of the decrease in the metabolite formed by CYP1A2 was larger than that by CYP2C9. mfa-quinoneimine 17-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 225-231 28520369-0 2012 Celecoxib Therapy and CYP2C9 Genotype Celecoxib is a nonsteroidal anti-inflammatory drug (NSAID) that is used in the management of osteoarthritis, rheumatoid arthritis, menstrual symptoms, and acute pain. Celecoxib 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 28520369-3 2012 The CYP2C9 gene encodes an enzyme involved in the metabolism of many drugs, and is one of the main enzymes that metabolizes and inactivates celecoxib. Celecoxib 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 33542643-2 2021 Due to rifampin"s strong induction of CYP2C9, most cases could not attain the target international normalized ratio (INR) despite warfarin dose escalation. Rifampin 7-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 28520369-6 2012 The FDA-approved drug label for celecoxib states: "patients who are known or suspected to be poor CYP2C9 metabolizers based on genotype or previous history/experience with other CYP2C9 substrates (such as warfarin, phenytoin) should be administered celecoxib with caution. Celecoxib 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 33542643-8 2021 The analysis revealed that the subject carries CYP2C9*3*3 and VKORC1-1639 (GA) mutations, classifying her as a slow metabolizer and, hence, highly warfarin-sensitive. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 28520369-6 2012 The FDA-approved drug label for celecoxib states: "patients who are known or suspected to be poor CYP2C9 metabolizers based on genotype or previous history/experience with other CYP2C9 substrates (such as warfarin, phenytoin) should be administered celecoxib with caution. Celecoxib 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 33093187-6 2021 Mitragynine was tested against CYP2C9 (diclofenac 4"-hydroxylation), CYP2D6 (dextromethorphan O-demethylation), and CYP3A (midazolam 1"-hydroxylation) activities in human liver microsomes (HLMs) and CYP3A activity in human intestinal microsomes (HIMs). mitragynine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 33498694-10 2021 Additionally, we describe that fluoxetine is a CYP2C9 substrate and a mechanism-based inhibitor of CYP2C19. Fluoxetine 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 33378980-5 2021 Polymorphisms in CYP2C9, SLC22A1, ABCB1, ABCC2, and APOC3 were related to olanzapine pharmacokinetic variability. Olanzapine 74-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 32860733-4 2021 In the U.S. cohort, VKORC1 and CYP2C9 variants were associated with lower warfarin dose (beta=-0.29, p<2.0x10-16 ; beta=-0.21, p=4.7x10-7 , respectively) whereas CYP4F2 and NQO1 variants were associated with higher dose (beta=0.10, p=2x10-4 ; beta=0.10, p=0.01, respectively). Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 32860733-7 2021 We confirmed the association of variants in VKORC1, CYP2C9, and CYP4F2 with warfarin dose in Latinos from U.S. and Brazil. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 33309749-5 2021 Furthermore, in contrast to imidazole derivative C38, the tert-butylimidazole derivative 20 is a poor inhibitor of CYP3A4, CYP2D6 and CYP2C9. tert-butylimidazole 58-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 33321412-5 2021 Knipholone was also the most effective in increasing the expression of CYP3A4 (8.47 +- 2.5 fold) and CYP2C9 (2.64 +- 0.3 fold) at 10 microM. knipholone 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 33321412-7 2021 In a CYP inhibition assay, the methanolic extract as well as the anthraquinones strongly inhibited the catalytic activity of CYP2C9 while, inhibition of CYP3A4 was weak. Anthraquinones 65-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 32813611-0 2021 Main contribution of UGT1A1 and CYP2C9 in the metabolism of UR-1102, a novel agent for the treatment of gout. UR-1102 60-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 31709648-0 2020 The presence of two reduced function variants in CYP2C9 influences the acute response to glipizide. Glipizide 89-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 33341937-0 2021 Hypoglycemia and glycemic control with glyburide in women with gestational diabetes and genetic variants of cytochrome P450 2C9 and/or OATP1B3. Glyburide 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-127 33341937-1 2021 Glyburide is mainly metabolized by the cytochrome P450 2C9 (CYP2C9) enzyme and enters the liver via the transporter OATP1B3. Glyburide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-58 33341937-1 2021 Glyburide is mainly metabolized by the cytochrome P450 2C9 (CYP2C9) enzyme and enters the liver via the transporter OATP1B3. Glyburide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 33341937-2 2021 The variants OATP1B3*4 (699 G>A; rs7311358) and CYP2C9*2 and *3 are known to have a significant influence on the hepatic uptake and metabolism of glyburide, with lower clearance than in the wild type. Glyburide 146-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 33341937-7 2021 In conclusion, the no function variants CYP2C9*3 and OATP1B3*4 are associated with a higher risk of hypoglycemia and a lower dose of glyburide in women with gestational diabetes treated with glyburide, which is consistent with the pharmacokinetic roles of both CYP2C9 and OATP1B3. Glyburide 133-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 33341937-7 2021 In conclusion, the no function variants CYP2C9*3 and OATP1B3*4 are associated with a higher risk of hypoglycemia and a lower dose of glyburide in women with gestational diabetes treated with glyburide, which is consistent with the pharmacokinetic roles of both CYP2C9 and OATP1B3. Glyburide 133-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 261-267 33341937-7 2021 In conclusion, the no function variants CYP2C9*3 and OATP1B3*4 are associated with a higher risk of hypoglycemia and a lower dose of glyburide in women with gestational diabetes treated with glyburide, which is consistent with the pharmacokinetic roles of both CYP2C9 and OATP1B3. Glyburide 191-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 31709648-8 2020 CONCLUSIONS: Carriers of two reduced function alleles in CYP2C9 may experience an increased insulin response to glipizide and be predisposed to a higher risk of hypoglycaemia, although no effect of genotype was seen in glucose-based measurements. Glipizide 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 31709648-8 2020 CONCLUSIONS: Carriers of two reduced function alleles in CYP2C9 may experience an increased insulin response to glipizide and be predisposed to a higher risk of hypoglycaemia, although no effect of genotype was seen in glucose-based measurements. Glucose 219-226 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 31709648-9 2020 Further studies are needed to clarify the utility of CYP2C9 genotyping to guide sulfonylurea treatment. Sulfonylurea Compounds 80-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 32279279-5 2020 Moreover, iloperidone attenuated the activity of CYP1A2 (Ki = 45 and 31 microM in microsomes and supersomes) and CYP2C19 via a mixed mechanism (Ki = 6.5 and 32 microM in microsomes and supersomes) but did not affect CYP2C9. iloperidone 10-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 216-222 32648321-12 2020 CONCLUSIONS: CYP1A1 and CYP2C9, UGT1A1, UGT1A7, UGT1A8 and UGT1A9, and MRP4 all played important roles in the metabolism and disposition of bavachin. bavachin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 33635755-14 2020 CBD is hydroxylated to 7-OH-CBD and 7-COOH-CBD by cytochrome P450 enzymes CYP3A4 and CYP2C9 in the liver and is excreted mainly in feces and less in urine. Cannabidiol 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 33635755-14 2020 CBD is hydroxylated to 7-OH-CBD and 7-COOH-CBD by cytochrome P450 enzymes CYP3A4 and CYP2C9 in the liver and is excreted mainly in feces and less in urine. 7-Hydroxycannabidiol 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 33635755-14 2020 CBD is hydroxylated to 7-OH-CBD and 7-COOH-CBD by cytochrome P450 enzymes CYP3A4 and CYP2C9 in the liver and is excreted mainly in feces and less in urine. 7-cooh-cbd 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 31319733-8 2020 DISCUSSION: The elevation in INR can be explained by the inhibition of CYP2C9 by cannabis use causing decreased metabolism of warfarin. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 32223485-3 2020 In addition, the enzyme kinetic parameters were calculated.Results: The results showed that the activity of CYP3A4, CYP2E1 and CYP2C9 was inhibited by CEP, with IC50 values of 16.29, 25.62 and 24.57 muM, respectively, but other CYP isoforms were not affected. cepharanthine 151-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 32279279-6 2020 Lurasidone moderately inhibited CYP1A2 (Ki = 12.6 and 15.5 microM in microsomes and supersomes), CYP2C9 (Ki = 18 and 3.5 microM in microsomes and supersomes) and CYP2C19 via a mixed mechanism (Ki = 18 and 18.4 microM in microsomes and supersomes), and CYP3A4 via a competitive mechanism (Ki = 29.4 and 9.1 microM in microsomes and supersomes). Lurasidone Hydrochloride 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 33350885-0 2020 Effect of CYP2C9 *11/*11 genotype on initial and long-term warfarin dose requirement and therapeutic response. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 32279279-9 2020 The obtained results indicate that metabolic/pharmacokinetic interactions with iloperidone (involving mainly CYP3A4 and CYP2D6) and possibly with lurasidone (involving CYP1A2, CYP2C9 or CYP2C19) may occur during combined therapy. Lurasidone Hydrochloride 146-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 32504053-3 2020 In a retrospective case-control study in European American patients with T2DM, we examined the potential association between CYP2C9 reduced-function variants and sulfonylurea-related hypoglycemia. Sulfonylurea Compounds 162-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 33350885-4 2020 This case report sheds further light on the clinical impact of CYP2C9 *11/*11 on warfarin dose requirements, short- and long-term treatment response and practical considerations for warfarin management in suspected carriers of rare variant CYP2C9 alleles. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 33350885-4 2020 This case report sheds further light on the clinical impact of CYP2C9 *11/*11 on warfarin dose requirements, short- and long-term treatment response and practical considerations for warfarin management in suspected carriers of rare variant CYP2C9 alleles. Warfarin 182-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 240-246 33262574-2 2020 Methods: The recombinant cytochrome P450 alleles protein of CYP2C9 and CYP3A4 expressed in insect baculovirus expression system were reacted with 10-500 microM sildenafil for 30 minutes at 37 C, and the reaction was terminated by cooling to -80 C immediately. Sildenafil Citrate 160-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 33180507-6 2020 Additionally, (S)-18 exhibits no apparent inhibitory effect on CYP isoforms except for a moderate effect on CYP2C9. (s)-18 14-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 33262574-0 2020 Functional Measurement of CYP2C9 and CYP3A4 Allelic Polymorphism on Sildenafil Metabolism. Sildenafil Citrate 68-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 33262574-9 2020 Conclusion: These findings were the first evaluation of all these infrequent CYP2C9 and CYP3A4 alleles for sildenafil metabolism; when treating people who carry these CYP2C9 and CYP3A4 variants, there should be more focus on the relation of dose intensity, side effects and therapeutic efficacy when administering sildenafil. Sildenafil Citrate 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 33262574-9 2020 Conclusion: These findings were the first evaluation of all these infrequent CYP2C9 and CYP3A4 alleles for sildenafil metabolism; when treating people who carry these CYP2C9 and CYP3A4 variants, there should be more focus on the relation of dose intensity, side effects and therapeutic efficacy when administering sildenafil. Sildenafil Citrate 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 33262574-9 2020 Conclusion: These findings were the first evaluation of all these infrequent CYP2C9 and CYP3A4 alleles for sildenafil metabolism; when treating people who carry these CYP2C9 and CYP3A4 variants, there should be more focus on the relation of dose intensity, side effects and therapeutic efficacy when administering sildenafil. Sildenafil Citrate 314-324 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 33262574-9 2020 Conclusion: These findings were the first evaluation of all these infrequent CYP2C9 and CYP3A4 alleles for sildenafil metabolism; when treating people who carry these CYP2C9 and CYP3A4 variants, there should be more focus on the relation of dose intensity, side effects and therapeutic efficacy when administering sildenafil. Sildenafil Citrate 314-324 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 33262574-10 2020 The study will provide fundamental data on effect of CYP2C9 and CYP3A4 allelic variation on sildenafil metabolism for further clinical research. Sildenafil Citrate 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 33240461-1 2020 CYP2C9 gene encodes an enzyme involved in the metabolism of a wide variety of drugs which include celecoxib. Celecoxib 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33330279-7 2020 We also analyzed the influence of exogenous factors on the level of 25(OH)D in children of the three study regions, as well as the relationship of the level of 25(OH)D with variants CYP2C9 * 2 (c.430C>T; R144C), CYP2C9 * 3 (c,1075A>C; I359L), CYP2D6 * 4 (1846G>A), CYP3A4 * 3 (c.1334T>C), and CYP3A4 * 1B (c.-392C>T) and rs731236, rs2228570 and rs1544410 in the VDR gene. 25(oh)d 160-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 33330279-14 2020 Conclusion: Exogenous factors (time of year, place of residence, and prophylactic administration of cholecalciferol), as well as endogenous factors (age and sex), play a determining role in the development of vitamin D deficiency and insufficiency; in contrast to genetic factors-polymorphic variants of the genes of xenobiotic phase 1 enzymes (CYP2C9, CYP2C19, CYP2D6, and CYP3A4) and the VDR gene-which do not play such role. Vitamin D 209-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 345-351 33181661-5 2020 Therefore, it is recommended to detect the genotype of CYP2C9 in patients and evaluate the interaction between the 2 drugs to adjust the warfarin dose. Warfarin 137-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 33147873-0 2020 Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling to Predict the Impact of CYP2C9 Genetic Polymorphisms, Co-Medication and Formulation on the Pharmacokinetics and Pharmacodynamics of Flurbiprofen. Flurbiprofen 196-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 33147873-3 2020 Flurbiprofen has absolute bioavailability of ~95% and linear pharmacokinetics in the dose range of 50-300 mg. Its absorption is considered variable and complex, often associated with double peak phenomena, and its pharmacokinetics are characterized by high inter-subject variability, mainly due to its metabolism by the polymorphic CYP2C9 (fmCYP2C9 >= 0.71). Flurbiprofen 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 332-338 32583677-1 2020 Aim: Our aim was to examine the effect of CYP2C9 and VKORC1 polymorphisms on warfarin dose requirements in Turkish patients. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 32583677-3 2020 Results: Patients with CYP2C9 *1/*1 and VKORC1 -1639 GG and GA genotypes required higher warfarin doses in comparison to wild type VKORC1 genotype. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 32583677-4 2020 Patients with CYP2C9 *1/*3 and VKORC1 -1639 GG genotypes simultaneously, required the lowest dose of warfarin (4.64 mg/day). Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32583677-5 2020 Patients with CYP2C9 *1/*1 and VKORC1 9041 AA genotype were found to require higher warfarin doses. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32583677-6 2020 Conclusion: Our results provide additional evidence to support the hypothesis that CYP2C9 *2, *3, VKORC1 9041 G > A polymorphisms explain considerable proportion of inter-individual variability in warfarin dose requirement. Warfarin 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 82-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 82-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 82-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Dronabinol 93-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Carbonic Acid 97-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Carbonic Acid 97-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-8 2020 In addition, this study showed significantly higher values in the ratio of Delta9-THC/Delta9-THC-COOH for the carriers of the CYP2C9 variants CYP2C9*2 and CYP2C9*3 compared with the carriers of the corresponding wild-type alleles. Carbonic Acid 97-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32712703-9 2020 Therefore, an impact of variations of the CYP2C9 gene on the interpretation of cannabinoid plasma concentrations in DUID cases should be considered. Cannabinoids 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 33138123-4 2020 AB-PINACA reversibly inhibited the metabolic activities of CYP2C8 (Ki, 16.9 microM), CYP2C9 (Ki, 6.7 microM), and CYP2C19 (Ki, 16.1 microM) and the transport activity of OAT3 (Ki, 8.3 microM). N-(1-amino-3-methyl-1-oxobutan-2-yl)-1-pentyl-1H-indazole-3-carboxamide 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 32882369-11 2020 Systemic 0.1 % terbinafine treatment in the Chip2 increased RHS expression of EGFR, increased apoptotic cells in the liver, downregulated liver albumin expression and upregulated CYP2C9 significantly (p < 0.05), acting as an effective hepatotoxic terbinafine control. Terbinafine 15-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 32712703-7 2020 We recognized significantly lower Delta9-THC-COOH concentrations for CYP2C9*3 (p = 0.001) and a trend of lower Delta9-THC-COOH concentrations for CYP2C9*2 which did not reach statistical significance (p = 0.068). Carbonic Acid 45-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 32712703-7 2020 We recognized significantly lower Delta9-THC-COOH concentrations for CYP2C9*3 (p = 0.001) and a trend of lower Delta9-THC-COOH concentrations for CYP2C9*2 which did not reach statistical significance (p = 0.068). delta9 34-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 32938720-0 2020 Structure of Cytochrome P450 2C9*2 in Complex with Losartan: Insights into the Effect of Genetic Polymorphism. Losartan 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-32 32938720-4 2020 The crystal structure of CYP2C9*2 in complex with an antihypertensive drug losartan was solved using X-ray crystallography at 3.1-A resolution. Losartan 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 32938720-5 2020 The Arg144Cys variation in the *2 complex disrupts the hydrogen-bonding interactions that were observed between the side chain of arginine and neighboring residues in the losartan complex of CYP2C9 and the wild-type (WT) ligand-free structure. arg144cys 4-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 32938720-5 2020 The Arg144Cys variation in the *2 complex disrupts the hydrogen-bonding interactions that were observed between the side chain of arginine and neighboring residues in the losartan complex of CYP2C9 and the wild-type (WT) ligand-free structure. Hydrogen 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 32938720-5 2020 The Arg144Cys variation in the *2 complex disrupts the hydrogen-bonding interactions that were observed between the side chain of arginine and neighboring residues in the losartan complex of CYP2C9 and the wild-type (WT) ligand-free structure. Arginine 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 32938720-5 2020 The Arg144Cys variation in the *2 complex disrupts the hydrogen-bonding interactions that were observed between the side chain of arginine and neighboring residues in the losartan complex of CYP2C9 and the wild-type (WT) ligand-free structure. Losartan 171-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 32938720-7 2020 The new structure revealed distinct interactions of losartan in the compact active site of CYP2C9*2 and differed in occupancy at the other binding sites previously identified in the WT-losartan complex. Losartan 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 32938720-8 2020 Furthermore, the binding studies in solution using losartan illustrated lower activity of the CYP2C9*2 compared with the WT. Losartan 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 32938720-9 2020 Together, the findings yield valuable insights into the decreased hydroxylation activity of losartan in patients carrying CYP2C9*2 allele and provide a useful framework to investigate the effect of a single-nucleotide polymorphism that leads to altered metabolism of diverse drug substrates. Losartan 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 32938720-11 2020 How the CYP2C9*2 variant induces altered drug metabolism is poorly understood given that the Arg144Cys variation is located far away from the active site. arg144cys 93-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 32763815-14 2020 Isozymes of CYP1A2, CYP2C9, UGT1A1, UGT1A6, UGT1A9, UGT2B7 and UGT2B15 were involved in phase I and phase II metabolism of osalmid. osalmide 123-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 32661014-6 2020 Our observations revealed that chrysin conjugates are strong inhibitors of certain biotransformation enzymes (e.g., CYP2C9) and transporters (e.g., OATP1B1, OATP1B3, OATP2B1, and BCRP) examined. chrysin 31-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 32628284-7 2020 Simultaneously, PXR-RXRalpha would activate the expression of CYP2C9; metabolic kinetics of ursolic acid in CYP metabolizing enzyme lysate of Caco2 cells and Caco2-PXR-RXR cells was studied and it was found that the Km values were 81.99 +- 44.32 and 60.05 +- 29.62 microg/ml, and Vmax values were 3.77 +- 0.86 and 3.41 +- 0.96 microg ml-1 min-1 , respectively. ursolic acid 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 33071782-0 2020 Comparison of CYP2C9 Activity in Ethiopian and Non-Ethiopian Jews Using Phenytoin as a Probe. Phenytoin 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 33071782-2 2020 The objective of the study was to compare CYP2C9 activity by using Phenytoin Metabolic Ratio (PMR) between Ethiopian and non-Ethiopian Jews. Phenytoin 67-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 32968106-4 2020 We studied modulation by microsomal membrane-bound and soluble cyt b5 of the hydroxylation of salicylic acid to gentisic acid and ROS release by CYP2C9 activity in human liver microsomes (HLMs) and by CYP2C9 baculosomes. Reactive Oxygen Species 130-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 32968106-5 2020 CYP2C9 accounts for nearly 75% of salicylic acid hydroxylation in HLMs at concentrations reached after usual aspirin doses. Salicylic Acid 34-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32968106-5 2020 CYP2C9 accounts for nearly 75% of salicylic acid hydroxylation in HLMs at concentrations reached after usual aspirin doses. Aspirin 109-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32968106-6 2020 The anti-cyt b5 antibody SC9513 largely inhibits the rate of salicylic acid hydroxylation by CYP2C9 in HLMs and CYP2C9 baculosomes, increasing the KM approximately threefold. sc9513 25-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32968106-6 2020 The anti-cyt b5 antibody SC9513 largely inhibits the rate of salicylic acid hydroxylation by CYP2C9 in HLMs and CYP2C9 baculosomes, increasing the KM approximately threefold. sc9513 25-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 32968106-6 2020 The anti-cyt b5 antibody SC9513 largely inhibits the rate of salicylic acid hydroxylation by CYP2C9 in HLMs and CYP2C9 baculosomes, increasing the KM approximately threefold. Salicylic Acid 61-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 33005200-4 2020 100 muM of kurarinone strongly inhibited more than 90% of UGT1A1, UGT1A6, CYP1A2, and CYP2C9. kurarinone 11-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 32863324-0 2020 Phenotypic analysis of human CYP2C9 polymorphisms using fluorine-substituted tolbutamide. Fluorine 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 32863324-0 2020 Phenotypic analysis of human CYP2C9 polymorphisms using fluorine-substituted tolbutamide. Tolbutamide 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 32863324-1 2020 To investigate the effect of fluorine substitution on tolbutamide (TB) hydroxylation catalyzed by CYP2C9, the hydroxylation of TB and its fluorinated derivative 3"-fluoro-tolbutamide (3"-F-TB) by recombinant human CYP2C9*1, CYP2C9*2, and CYP2C9*3 was analyzed. Fluorine 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 32863324-1 2020 To investigate the effect of fluorine substitution on tolbutamide (TB) hydroxylation catalyzed by CYP2C9, the hydroxylation of TB and its fluorinated derivative 3"-fluoro-tolbutamide (3"-F-TB) by recombinant human CYP2C9*1, CYP2C9*2, and CYP2C9*3 was analyzed. Tolbutamide 54-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 32863324-1 2020 To investigate the effect of fluorine substitution on tolbutamide (TB) hydroxylation catalyzed by CYP2C9, the hydroxylation of TB and its fluorinated derivative 3"-fluoro-tolbutamide (3"-F-TB) by recombinant human CYP2C9*1, CYP2C9*2, and CYP2C9*3 was analyzed. Tolbutamide 67-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 32591416-10 2020 SIGNIFICANCE STATEMENT: Robust kinetic parameters were determined for the reversible and time-dependent inhibition of CYP2C9, CYP2D6 and CYP3A4/5 activities in HLMs by major component isoquinoline alkaloids contained in the botanical natural product (NP) goldenseal. isoquinoline 184-196 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 32863324-1 2020 To investigate the effect of fluorine substitution on tolbutamide (TB) hydroxylation catalyzed by CYP2C9, the hydroxylation of TB and its fluorinated derivative 3"-fluoro-tolbutamide (3"-F-TB) by recombinant human CYP2C9*1, CYP2C9*2, and CYP2C9*3 was analyzed. Tolbutamide 127-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 32863324-1 2020 To investigate the effect of fluorine substitution on tolbutamide (TB) hydroxylation catalyzed by CYP2C9, the hydroxylation of TB and its fluorinated derivative 3"-fluoro-tolbutamide (3"-F-TB) by recombinant human CYP2C9*1, CYP2C9*2, and CYP2C9*3 was analyzed. 3"-f-tb 184-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 32661920-1 2020 Zolpidem, a widely prescribed hypnotic agent, is extensively metabolized by cytochrome P450 (CYP) 3A4, and CYP2C9, CYP1A2 and CYP2D6 are also involved in the metabolism of zolpidem. Zolpidem 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 32661920-1 2020 Zolpidem, a widely prescribed hypnotic agent, is extensively metabolized by cytochrome P450 (CYP) 3A4, and CYP2C9, CYP1A2 and CYP2D6 are also involved in the metabolism of zolpidem. Zolpidem 172-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 32628284-8 2020 However, in human CYP metabolizing recombinase, we found that both CYP2C9 and CYP34A were involved in the metabolism of ursolic acid. ursolic acid 120-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 32729746-4 2020 Fluvastatin is metabolized by CYP2C9, whereas pravastatin, rosuvastatin, and pitavastatin are unaffected by inhibition by either CYP. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 32338345-7 2020 CONCLUSIONS: Co-administration of apalutamide with CYP3A4, CYP2C19, CYP2C9, P-gp, BCRP or OATP1B1 substrates may cause loss of activity for these medications. apalutamide 34-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 32216088-0 2020 Associations of CYP2C9 and CYP2C19 pharmacogenetic variation with phenytoin-induced cutaneous adverse drug reactions. Phenytoin 66-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32216088-1 2020 The role of CYP2C9 and CYP2C19 genetic variation in risk for phenytoin-induced cutaneous adverse drug events is not well understood independently of the HLA-B*15:02 risk allele. Phenytoin 61-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 32216088-9 2020 Genetic variation reducing CYP2C9 metabolic activity may increase risk for phenytoin-induced cutaneous adverse events in the absence of the HLA-B*15:02 risk allele. Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 32270628-0 2020 Impact of CYP2C9-Interacting Drugs on Warfarin Pharmacogenomics. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 32270628-4 2020 Study participants predicted to be sensitive responders to warfarin based on CYP2C9 and VKORC1 genotypes, had significantly greater international normalized ratio (INR) variability over time. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 32773350-0 2020 CYP2 C9 polymorphism among patients with oral squamous cell carcinoma and its role in altering the metabolism of benzo[a]pyrene. Benzo(a)pyrene 113-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-7 33967488-0 2020 How genetic variation was analyzed in phenytoin-induced gingival enlargement using single-nucleotide polymorphism of candidate gene CYP2C9? Phenytoin 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 33967488-12 2020 However, larger scale genome-wide study has to be carried out to confirm one of the etiopathogenesis as mutation of the CYP2C9 gene, in phenytoin-induced gingival overgrowth. Phenytoin 136-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 32893731-0 2020 Effects of rare CYP2C9 alleles on stable warfarin doses in Chinese Han patients with atrial fibrillation. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32893731-2 2020 Here, the role of rare CYP2C9 alleles on warfarin doses in Chinese Han patients was investigated. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 32893731-7 2020 Inclusion of the rare CYP2C9 alleles in the prediction model added an extra 3.7% warfarin dose predictive power. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 32893731-8 2020 Conclusion: CYP2C9*13, *16 and *60 was associated with lower stable warfarin doses in Chinese patients. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 32893731-9 2020 The algorithm including rare CYP2C9 alleles tends to more accurately predict stable warfarin doses. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 32583524-8 2020 We found that orbitazine was metabolized by CYP3A4, CYP2C9 and CYP2D6 in a human liver microsomes incubation system. orbitazine 14-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32773350-2 2020 We also investigated the interaction between CYP2 C9 wild type and the polymorphic variants with benzo[a]pyrene by using molecular docking analysis. Benzo(a)pyrene 97-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-52 32773350-5 2020 Molecular docking was then carried out to determine the interaction of CYP2 C9*1, CYP2 C9*2, and CYP2 C9*3 with benzo[a]pyrene. Benzo(a)pyrene 112-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-78 32773350-5 2020 Molecular docking was then carried out to determine the interaction of CYP2 C9*1, CYP2 C9*2, and CYP2 C9*3 with benzo[a]pyrene. Benzo(a)pyrene 112-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-89 32773350-5 2020 Molecular docking was then carried out to determine the interaction of CYP2 C9*1, CYP2 C9*2, and CYP2 C9*3 with benzo[a]pyrene. Benzo(a)pyrene 112-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-89 32773350-8 2020 The docking analysis showed benzo[a]pyrene to bind specifically to the altered single nucleotide catalytic site in the polymorphic CYP2 C9*3 enzyme. Benzo(a)pyrene 28-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-138 32773350-10 2020 The modified docking of CYP2 C9*3 with benzo[a]pyrene signifies altered metabolism in vivo. Benzo(a)pyrene 39-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-31 32129697-5 2020 Rucaparib reversibly inhibited CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3As (IC50, 3.55, 12.9, 5.42, 41.6, and 17.2-22.9 microM [2 substrates], respectively), but not CYP2B6 or CYP2C8 (>190 microM). rucaparib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 32385545-0 2020 Influence of CYP2C9 and CYP2A6 on plasma concentrations of valproic acid: a meta-analysis. Valproic Acid 59-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 33194861-3 2020 Numerous genes have been implicated in warfarin pharmacogenetics, with genes encoding CYP2C9 and VKORC1 shown to be the most important determinants of drug dosage requirements. Warfarin 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 33194861-6 2020 LEARNING POINTS: Genetic warfarin resistance is rare and mainly associated with two genes encoding CYP2C9 and VKORC1.In addition to dietary counselling and drug compliance, options in warfarin-resistant patients include increasing the warfarin dose, which carries a risk of bleeding complications, or switching to novel oral anticoagulants, which increases the risk of prosthetic valve thrombosis.We replaced a metallic valve with a bioprosthetic valve, which is the first time this has been documented in a patient with warfarin resistance. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 33194861-6 2020 LEARNING POINTS: Genetic warfarin resistance is rare and mainly associated with two genes encoding CYP2C9 and VKORC1.In addition to dietary counselling and drug compliance, options in warfarin-resistant patients include increasing the warfarin dose, which carries a risk of bleeding complications, or switching to novel oral anticoagulants, which increases the risk of prosthetic valve thrombosis.We replaced a metallic valve with a bioprosthetic valve, which is the first time this has been documented in a patient with warfarin resistance. Warfarin 184-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 33194861-6 2020 LEARNING POINTS: Genetic warfarin resistance is rare and mainly associated with two genes encoding CYP2C9 and VKORC1.In addition to dietary counselling and drug compliance, options in warfarin-resistant patients include increasing the warfarin dose, which carries a risk of bleeding complications, or switching to novel oral anticoagulants, which increases the risk of prosthetic valve thrombosis.We replaced a metallic valve with a bioprosthetic valve, which is the first time this has been documented in a patient with warfarin resistance. Warfarin 184-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 33194861-6 2020 LEARNING POINTS: Genetic warfarin resistance is rare and mainly associated with two genes encoding CYP2C9 and VKORC1.In addition to dietary counselling and drug compliance, options in warfarin-resistant patients include increasing the warfarin dose, which carries a risk of bleeding complications, or switching to novel oral anticoagulants, which increases the risk of prosthetic valve thrombosis.We replaced a metallic valve with a bioprosthetic valve, which is the first time this has been documented in a patient with warfarin resistance. Warfarin 184-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 32601017-0 2020 Inhibitory effects of sesamin on CYP2C9-dependent 7-hydroxylation of S-warfarin. sesamin 22-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 32601017-0 2020 Inhibitory effects of sesamin on CYP2C9-dependent 7-hydroxylation of S-warfarin. Warfarin 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 32601017-2 2020 This finding suggests that sesamin predominantly binds to CYP2C9 at another site for which it has a higher affinity than its affinity for the active site, thereby inhibiting the activity of CYP2C9 non-competitively. sesamin 27-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 32601017-2 2020 This finding suggests that sesamin predominantly binds to CYP2C9 at another site for which it has a higher affinity than its affinity for the active site, thereby inhibiting the activity of CYP2C9 non-competitively. sesamin 27-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 32601017-4 2020 In addition, the recombinant CYP2C9-dependent 7-hydroxylation activity of S-warfarin was competitively inhibited by sesamin with a Ki value of 13.1 muM. Warfarin 74-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 32601017-4 2020 In addition, the recombinant CYP2C9-dependent 7-hydroxylation activity of S-warfarin was competitively inhibited by sesamin with a Ki value of 13.1 muM. sesamin 116-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 32601017-5 2020 These results are consistent with the fact that sesamin is a good substrate of CYP2C9, and its activity follows Michaelis-Menten kinetics. sesamin 48-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 32868386-0 2020 Impact of CYP2C19 and CYP2C9 gene polymorphisms on sodium valproate plasma concentration in patients with epilepsy. Valproic Acid 51-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 32868386-1 2020 BACKGROUND: Valproic acid (VPA) is a broad spectrum anticonvulsant drug, which could be partially metabolised by cytochrome P450 (CYP) 2C9 and 2C19 enzymes. Valproic Acid 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-147 32868386-1 2020 BACKGROUND: Valproic acid (VPA) is a broad spectrum anticonvulsant drug, which could be partially metabolised by cytochrome P450 (CYP) 2C9 and 2C19 enzymes. Valproic Acid 27-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-147 32868386-2 2020 This study was designed to investigate the relationship between CYP2C19 and CYP2C9 gene polymorphisms and the plasma concentrations of VPA in subjects with epilepsy. Valproic Acid 135-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 32868386-10 2020 RESULTS: The genetic frequencies of CYP2C19*2, CYP2C19*3 and CYP2C9*13 were 33.1%, 3.0% and 5.4%, respectively, among patients with epilepsy from Yunnan province, China who used VPA therapy. Valproic Acid 178-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 32531309-2 2020 The dose of warfarin varies significantly within populations, and the dose is closely related to the genetic polymorphisms of the CYP2C9 and VKORC1 genes. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 32531309-3 2020 In this study, a new CYP2C9 nonsynonymous mutation (8576C > T) was detected after the genetic screening of 162 patients took warfarin. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 32809952-4 2020 The k e of warfarin given a cytochrome P450 2C9 (CYP2C9) genotype ranged from 0.0189 to 0.0075 h-1. Warfarin 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-47 32809952-4 2020 The k e of warfarin given a cytochrome P450 2C9 (CYP2C9) genotype ranged from 0.0189 to 0.0075 h-1. Warfarin 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 32125665-4 2020 RESULTS: Finerenone caused direct inhibitory effects on CYP activities in vitro in the rank order CYP2C8, CYP1A1 > CYP3A4 > CYP2C9 and CYP2C19, but not on other major CYP isoforms. finerenone 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 32125665-6 2020 The major metabolites of finerenone demonstrated minor reversible inhibition of CYP1A1, CYP2C9 and CYP3A4 with no hint of time-dependent inhibition of any CYP isoform. finerenone 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 32385545-2 2020 Specifically, CYP2C9 and CYP2A6 are the main enzymes responsible for VPA metabolism. Valproic Acid 69-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32510242-2 2020 Genetic polymorphisms in the metabolism of phenytoin, particularly CYP2C9, are strongly associated with increased plasma concentrations and can result in toxicity. Phenytoin 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 32385545-3 2020 However, the correlation between plasma VPA concentrations and CYP2C9 and CYP2A6 gene variations is uncertain. Valproic Acid 40-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 32510242-4 2020 Recent pharmacogenomic studies show genetic polymorphisms in CYP2C9, as well as HLA alleles, are significantly associated with phenytoin-related SCAR. Phenytoin 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 32510242-5 2020 AREAS COVERED: Updated pharmacogenomic information of CYP2C9 variants and HLA alleles involved in phenytoin-associated cutaneous adverse drug reactions (cADRs) are discussed in this article. Phenytoin 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 32385545-4 2020 This meta-analysis aimed to investigate the relationship between CYP2C9 and CYP2A6 variants and plasma concentrations of VPA. Valproic Acid 121-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 32510242-6 2020 EXPERT OPINION: CYP2C9*3 has been identified as the most significant genetic variant associated with increased phenytoin concentrations and adverse events. Phenytoin 111-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32385545-10 2020 We found that CYP2C9*3 was associated with standardized plasma VPA concentration; *3 allele carriers had a 0.70-mug/mL higher concentration per mg/kg than non-carriers (95% CI 0.25, 1.15; P = 0.002). Valproic Acid 63-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32510242-7 2020 Recent pharmacogenomic findings reveal that CYP2C9*3 and HLA alleles, i.e. HLA-B*15:02, HLA-B*13:01, and HLA-B*51:01, are important genetic variants in the occurrence of phenytoin-induced cADRs or SCAR. Phenytoin 170-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 32385545-12 2020 CONCLUSION: This meta-analysis demonstrated that CYP2C9*3 and CYP2A6*4 genetic variants affect plasma VPA concentrations. Valproic Acid 102-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 32238050-1 2020 Osthol, a pharmacologically active ingredient in various traditional Chinese medicines, is predominantly metabolized by CYP2C9. osthol 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 32423989-1 2020 CYP2C9 is a major form of human liver cytochrome P450 that is responsible for the oxidative metabolism of several widely used low therapeutic index drugs, including (S)-warfarin and phenytoin. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32423989-1 2020 CYP2C9 is a major form of human liver cytochrome P450 that is responsible for the oxidative metabolism of several widely used low therapeutic index drugs, including (S)-warfarin and phenytoin. Phenytoin 182-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32423989-7 2020 Steady-state kinetic analyses revealed that the relative catalytic efficiency ratios of (S)-warfarin metabolism by the P279T and N218I variants were 87% and 24%, respectively, of wildtype CYP2C9 protein. Warfarin 88-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 32294527-1 2020 Epoxyeicosatrienoic acids (EETs) are synthetized from arachidonic acid by the action of members of the CYP2C and CYP2J subfamilies of cytochrome P450 (CYPs). epoxyeicosatrienoic acids 0-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-108 32294527-1 2020 Epoxyeicosatrienoic acids (EETs) are synthetized from arachidonic acid by the action of members of the CYP2C and CYP2J subfamilies of cytochrome P450 (CYPs). eets 27-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-108 32294527-1 2020 Epoxyeicosatrienoic acids (EETs) are synthetized from arachidonic acid by the action of members of the CYP2C and CYP2J subfamilies of cytochrome P450 (CYPs). Arachidonic Acid 54-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-108 32238050-0 2020 Osthole inhibited the activity of CYP2C9 in human liver microsomes and influenced indomethacin pharmacokinetics in rats. osthol 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 32700644-0 2020 Prediction of the impact of CYP2C9 and VKORC1 genotypes on warfarin-sorafenib interactions in whites and Asians. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 32700644-0 2020 Prediction of the impact of CYP2C9 and VKORC1 genotypes on warfarin-sorafenib interactions in whites and Asians. Sorafenib 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 32700644-1 2020 Aim: To predict the impact of the different CYP2C9 and VKORC1 genotypes on warfarin-sorafenib interactions in whites and Asians. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 32700644-1 2020 Aim: To predict the impact of the different CYP2C9 and VKORC1 genotypes on warfarin-sorafenib interactions in whites and Asians. Sorafenib 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 32238050-4 2020 Interestingly, we found that osthole inhibits the activity of CYP2C11 in a non-competitive manner in RLMs, while inhibits CYP2C9 activity in a competitive manner in pooled HLMs. osthol 29-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 32700644-3 2020 Results: INRs were predicted to be 2.0-2.1- versus 1.8-1.9-times higher in the presence of sorafenib in the CYP2C9 (*1/*1 vs *1/*3) groups than those for warfarin alone in both whites and Asians. Sorafenib 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 32238050-5 2020 Then, the effects of CYP2C9*1/*2/*3 allele on the pharmacokinetics of osthole were identified. osthol 70-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 174-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 174-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 174-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 174-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 174-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. Indomethacin 225-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. Indomethacin 225-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. Indomethacin 225-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. Indomethacin 225-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. Indomethacin 225-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 271-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 271-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 271-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 271-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32238050-6 2020 In human CYP2C9 isoform, the Ki value of 21.93 muM (CYP2C9*1), 18.10 muM (CYP2C9*2), 13.12 muM (CYP2C9*3) indicate that there are individual differences in the inhibition of osthole on CYP2C9 activity.We investigated how the indomethacin pharmacokinetics was affected by osthole in SD rat. osthol 271-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 32765265-6 2020 The effect of genetic polymorphisms known to influence dose response of warfarin (CYP2C9, VKORC1) were implemented into the model by modifying warfarin clearance (CYP2C9 *1: 0.2 L/h; *2: 0.14 L/h, *3: 0.04 L/h) and the concentration of available vitamin K (VKORC1 GA: -22% from normal vitamin K concentration; AA: -44% from normal vitamin K concentration). Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 32754031-9 2020 By means of stepwise multiple regression analysis, we established a new elder warfarin dosing algorithm (R2=0.3714) containing height, creatinine, amiodarone usage, CYP2C9 (*1*2 or *1*3), and VKORC1 (GA or GG) genotypes. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 32754031-12 2020 Polymorphism of CYP2C9 and VKORC1 obviously affected warfarin stable dose of the elder Han-Chinese. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32765265-6 2020 The effect of genetic polymorphisms known to influence dose response of warfarin (CYP2C9, VKORC1) were implemented into the model by modifying warfarin clearance (CYP2C9 *1: 0.2 L/h; *2: 0.14 L/h, *3: 0.04 L/h) and the concentration of available vitamin K (VKORC1 GA: -22% from normal vitamin K concentration; AA: -44% from normal vitamin K concentration). Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 32765265-6 2020 The effect of genetic polymorphisms known to influence dose response of warfarin (CYP2C9, VKORC1) were implemented into the model by modifying warfarin clearance (CYP2C9 *1: 0.2 L/h; *2: 0.14 L/h, *3: 0.04 L/h) and the concentration of available vitamin K (VKORC1 GA: -22% from normal vitamin K concentration; AA: -44% from normal vitamin K concentration). Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 32765265-6 2020 The effect of genetic polymorphisms known to influence dose response of warfarin (CYP2C9, VKORC1) were implemented into the model by modifying warfarin clearance (CYP2C9 *1: 0.2 L/h; *2: 0.14 L/h, *3: 0.04 L/h) and the concentration of available vitamin K (VKORC1 GA: -22% from normal vitamin K concentration; AA: -44% from normal vitamin K concentration). Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 33680011-0 2020 The Contribution of VKORC1 and CYP2C9 Genetic Polymorphisms and Patients" Demographic Characteristics with Warfarin Maintenance Doses: A Suggested Warfarin Dosing Algorithm. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 32276000-4 2020 The CYP2C9 genomic region holding important warfarin drug-metabolizing SNPs was sequenced from 159 individuals belong from five major ethnic groups of Pakistani population. Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 32276000-9 2020 Several SNPs, including the previously reported warfarin associated variants, i.e. rs2860905, rs1799853 (CYP2C9*2) and rs72558189 (CYP2C9*14) were detected in these population groups with diverse frequencies. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 32276000-12 2020 The pharmacogeneticsassessment of the CYP2C9 genetic variations identified in current study may important to test against the warfarin efficacy for different ethnicity of Pakistani population. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 33680011-0 2020 The Contribution of VKORC1 and CYP2C9 Genetic Polymorphisms and Patients" Demographic Characteristics with Warfarin Maintenance Doses: A Suggested Warfarin Dosing Algorithm. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 33680011-6 2020 The patients who carried the A allele at position -1639 VKORC1 and the variants CYP2C9 *2 and *3 required a significantly lower daily mean warfarin dosage (P = 0.001). Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 33680011-8 2020 This study showed that in the heart valve replacement patients considering VKORC1 and CYP2C9 polymorphisms beside demographic characteristics such as age will be helpful in pre-treatment dosing of warfarin which in turn reduces the complications associated with inappropriate warfarin dosing. Warfarin 197-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 32380173-7 2020 The warfarin sensitivity allele recommendations in this report incorporate the previous CYP2C9 allele recommendations and additional genes and alleles that are specific to warfarin sensitivity testing. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 33680011-8 2020 This study showed that in the heart valve replacement patients considering VKORC1 and CYP2C9 polymorphisms beside demographic characteristics such as age will be helpful in pre-treatment dosing of warfarin which in turn reduces the complications associated with inappropriate warfarin dosing. Warfarin 276-284 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 33612460-0 2020 Pharmacogenetics of sulfonylurea: Presence of CYP2C9*2, CYP2C9*3 and a novel allele, CYP2C9*61, in Type 2 diabetes patients under sulfonylurea therapy. Sulfonylurea Compounds 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 32586526-0 2020 Variants in ADRB1 and CYP2C9: Association with Response to Atenolol and Losartan in Marfan Syndrome. Atenolol 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 32586526-0 2020 Variants in ADRB1 and CYP2C9: Association with Response to Atenolol and Losartan in Marfan Syndrome. Losartan 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 32586526-2 2020 STUDY DESIGN: In a subset of 250 white, non-Hispanic participants with Marfan syndrome in a prior randomized trial of atenolol vs losartan, the common variants rs1801252 and rs1801253 in ADRB1 and rs1799853 and rs1057910 in CYP2C9 were analyzed. Atenolol 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-230 33612460-0 2020 Pharmacogenetics of sulfonylurea: Presence of CYP2C9*2, CYP2C9*3 and a novel allele, CYP2C9*61, in Type 2 diabetes patients under sulfonylurea therapy. Sulfonylurea Compounds 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33583789-6 2020 Furthermore, we demonstrated that Paeoniflorin and Amygdalin had obvious inhibiting effects on CYP2C9 and CYP2E1. peoniflorin 34-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 33612460-0 2020 Pharmacogenetics of sulfonylurea: Presence of CYP2C9*2, CYP2C9*3 and a novel allele, CYP2C9*61, in Type 2 diabetes patients under sulfonylurea therapy. Sulfonylurea Compounds 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 33583789-6 2020 Furthermore, we demonstrated that Paeoniflorin and Amygdalin had obvious inhibiting effects on CYP2C9 and CYP2E1. Amygdalin 51-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 33612460-1 2020 CYP2C9 is an important member of the cytochrome P450 gene family involved in the metabolism of 15% of the drugs including an oral antidiabetic agent sulfonylurea. Sulfonylurea Compounds 149-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 33612460-2 2020 This study aims to investigate the frequency of CYP2C9*2 and CYP2C9*3 alleles of the gene in the sulfonylurea treated diabetic subjects in Pakistan. Sulfonylurea Compounds 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 33612460-2 2020 This study aims to investigate the frequency of CYP2C9*2 and CYP2C9*3 alleles of the gene in the sulfonylurea treated diabetic subjects in Pakistan. Sulfonylurea Compounds 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). tert-Butylhydroperoxide 157-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32575674-0 2020 rs622342 in SLC22A1, CYP2C9*2 and CYP2C9*3 and Glycemic Response in Individuals with Type 2 Diabetes Mellitus Receiving Metformin/Sulfonylurea Combination Therapy: 6-Month Follow-Up Study. Metformin 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 32575674-0 2020 rs622342 in SLC22A1, CYP2C9*2 and CYP2C9*3 and Glycemic Response in Individuals with Type 2 Diabetes Mellitus Receiving Metformin/Sulfonylurea Combination Therapy: 6-Month Follow-Up Study. Sulfonylurea Compounds 130-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 32575674-11 2020 CONCLUSION: The combination of metformin/sulfonylurea therapy led to the maximum glycemic control in individuals with T2DM carrying AA or AC genotypes in SLC22A1 and *1*3 in CYP2C9. Metformin 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 32575674-11 2020 CONCLUSION: The combination of metformin/sulfonylurea therapy led to the maximum glycemic control in individuals with T2DM carrying AA or AC genotypes in SLC22A1 and *1*3 in CYP2C9. Sulfonylurea Compounds 41-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 174-180 32606720-0 2020 The Effect of CYP2C9 Genotype Variants in Type 2 Diabetes on the Pharmacological Effectiveness of Sulfonylureas, Diabetic Retinopathy, and Nephropathy. Sulfonylurea Compounds 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 214-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 214-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 341-347 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). OSS 146-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). Hydrogen Peroxide 151-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32606720-10 2020 The findings also showed that the plasma level of sulfonylureas (i.e., glibenclamide and gliclazide) was the highest in the patients with the CYP2C9*3 allele. Sulfonylurea Compounds 50-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32606720-10 2020 The findings also showed that the plasma level of sulfonylureas (i.e., glibenclamide and gliclazide) was the highest in the patients with the CYP2C9*3 allele. Glyburide 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32606720-10 2020 The findings also showed that the plasma level of sulfonylureas (i.e., glibenclamide and gliclazide) was the highest in the patients with the CYP2C9*3 allele. Gliclazide 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 32527038-8 2020 There was a trend for an association of phenprocoumon-risk profiles (low VKORC1 or CYP2C9 activity) with phenprocoumon-suspecting ADRs (p = 0.052). Phenprocoumon 40-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 32527038-8 2020 There was a trend for an association of phenprocoumon-risk profiles (low VKORC1 or CYP2C9 activity) with phenprocoumon-suspecting ADRs (p = 0.052). Phenprocoumon 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 32279544-3 2020 The aim of this study was to determine whether the presence of certain allelic variants in genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in Mexican children with solid tumours treated with ifosfamide.Objective: The aim of this study was to determine whether the presence of certain allelic variants of genes CYP2B6, CYP2C9, CYP3A4 and CYP3A5 increases the risk of toxicity in children with solid tumours treated with ifosfamide.Materials and methods: A total of 131 DNA samples were genotyped by real-time polymerase chain reaction (RT-PCR) using TaqMan probes. Ifosfamide 442-452 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 32193358-7 2020 A competitive inhibition between PZQ enantiomers for CYP2C9, 2C19, 3A4 and 3A5 was revealed. Praziquantel 33-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). tert-Butylhydroperoxide 183-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). cuooh 196-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). phenyliodosodiacetate 203-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). 3-aminoisobutyric acid 227-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). (bis(trifluoroacetoxy)iodo)benzene 238-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32238418-5 2020 Here, we investigated the ability of the major human drug-metabolizing P450s, namely CYP3A4, CYP2C9, CYP2C19, CYP2D6 and CYP1A2, to use different OSs: H2O2; tert-butyl hydroperoxide (tert-BuOOH); CuOOH; (diacetoxyiodo)benzene (BAIB); and bis(trifluoroacetoxy)iodobenzene (F-BAIB). 3-aminoisobutyric acid 273-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32453717-0 2020 CYP2C9, a Metabolic CYP450s Enzyme, Plays Critical Roles in Activating Ellagic Acid in Human Intestinal Epithelial Cells. Ellagic Acid 71-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32453717-9 2020 EA treatment at different concentrations (10 mug/mL and 50 mug/mL) remarkably decreased cell viability of HIEC cells expressing CYP2C9 compared to the untreated control (p<0.01), in a concentration-dependent and time-dependent manner. Ellagic Acid 0-2 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 32453717-10 2020 Expression of CYP2C9 significantly increased the apoptosis rate of HIEC cells treated with EA compared to that in HIEC cells without any CYP450s expression (p<0.01). Ellagic Acid 91-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 32453717-11 2020 The clearance rate of EA in CYP2B6-expressing (p<0.05) and CYP2C9-expressing (p<0.001) HIEC cell models was remarkably reduced after 120 min. Ellagic Acid 22-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 32453717-12 2020 CONCLUSIONS Ellagic acid was effectively activated by CYP2C9 in HIEC cells and caused cytotoxicity and apoptosis of HIEC cells. Ellagic Acid 12-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 32453717-13 2020 Therefore, CYP2C9 is main metabolic enzyme of EA when compared to other CYP450 HIEC cell models. Ellagic Acid 46-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 32457604-10 2020 Conclusions: Decreased function alleles of CYP2C9 were highly predictive of vestibular-cerebellar ADRs to phenytoin in our study (p = 0.001). Phenytoin 106-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 32546963-2 2020 The in vitro inhibition studies suggested that apatinib exerted potent inhibition on CYP3A4 and CYP2C9. apatinib 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 32546963-12 2020 Owing to the risk of pharmacokinetic drug-drug interactions based on CYP3A4/CYP2C9 inhibition by apatinib, caution is advised in the concurrent use of apatinib with either CYP2C9 or CYP3A4 substrates. apatinib 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 32457621-2 2020 We aimed to investigate frequency and concordance on CYP2D6, CYP2C19, and CYP2C9 (CYP2D6/2C19/2C9)-mediated potential DDIs at the Lifelines cohort and linked data from the pharmacy database IADB.nl. Didanosine 118-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 32457621-2 2020 We aimed to investigate frequency and concordance on CYP2D6, CYP2C19, and CYP2C9 (CYP2D6/2C19/2C9)-mediated potential DDIs at the Lifelines cohort and linked data from the pharmacy database IADB.nl. Didanosine 118-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-97 32406758-3 2020 CYP2C9 is one of the most abundant CYP enzymes and responsible for the metabolism of over 15% clinical drugs, including oral sulfonylurea hypoglycemics, nonsteroidal anti-inflammatory agents, selective cyclooxygenase-2 inhibitors, antiepileptics, angiotensin II receptor inhibitors and anticoagulants. Sulfonylurea Compounds 125-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30277905-7 2020 Edoxaban is superior to warfarin in patients with increased risk of bleeding with warfarin because of variants in CYP2C9 and VKORC1 genes. edoxaban 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 30277905-7 2020 Edoxaban is superior to warfarin in patients with increased risk of bleeding with warfarin because of variants in CYP2C9 and VKORC1 genes. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 31646624-0 2020 HLA-B*51:01 and CYP2C9*3 are risk factors for phenytoin-induced eruption in the Japanese population: analysis of data from the Biobank Japan Project. Phenytoin 46-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 31646624-1 2020 CYP2C9*3 and HLA-B alleles are reportedly associated with phenytoin-induced eruption in some East Asian populations; however, this finding is not readily applicable to the Japanese population. Phenytoin 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31646624-5 2020 CYP2C9*3 carrier status was significantly associated with phenytoin-induced eruption (P = 0.0022, odds ratio 7.05, 95% confidence interval 2.44-20.4). Phenytoin 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31646624-8 2020 CYP2C9*3 and HLA-B*51:01 allele carrier statuses are significantly associated with phenytoin-induced eruption; thus, checking this carrier status before prescription would decrease it in clinical practice. Phenytoin 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32406758-4 2020 Diclofenac (4"-hydroxylase) and tolbutamide (methylhydroxylation) are widely used as probe substrates for CYP2C9. Diclofenac 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 32406758-4 2020 Diclofenac (4"-hydroxylase) and tolbutamide (methylhydroxylation) are widely used as probe substrates for CYP2C9. Tolbutamide 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 32274641-5 2020 This is an observational nested case-control study that was conducted on a cohort of Qatari patients treated with warfarin with previously identified genotype for the CYP2C9, VKORC1, and CYP2F4. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 31938857-0 2020 Phenytoin intoxication associated with omeprazole administration in a child with defective CYP2C9. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 31938857-0 2020 Phenytoin intoxication associated with omeprazole administration in a child with defective CYP2C9. Omeprazole 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 31938857-2 2020 Phenytoin is a commonly used drug, and its metabolism is mediated by a specific cytochrome-P450 isoform, CYP2C9, which is encoded by a polymorphic gene. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 31938857-4 2020 Here we describe a case of phenytoin intoxication in a child with defective CYP2C9, after omeprazole administration. Phenytoin 27-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 31938857-4 2020 Here we describe a case of phenytoin intoxication in a child with defective CYP2C9, after omeprazole administration. Omeprazole 90-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 31848875-3 2020 In a recent study in patients awaiting surgery, following warfarin cessation the INR declined slower over time in those with two CYP2C9 variant alleles, increasing age, weight and number of comorbidities and that INR decline was faster in those with higher maintenance INR value. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 31724188-7 2020 Several case reports show cannabis may inhibit the metabolism of warfarin because of CYP2C9 interactions, resulting in increased plasma concentrations, increased international normalized ratio , and risk of bleeding. Warfarin 65-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 32390827-12 2020 CYP2C9 *2 or *3 carriers were associated with increased clearance of CPA. Cyclophosphamide 69-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 32390827-18 2020 CYP3A5 and CYP2C9 genotypes, BSA, BMI, and CPA dosage regimen influence PK of CPA. Cyclophosphamide 43-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 32390827-18 2020 CYP3A5 and CYP2C9 genotypes, BSA, BMI, and CPA dosage regimen influence PK of CPA. Cyclophosphamide 78-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 32290339-0 2020 Strong and Selective Inhibitory Effects of the Biflavonoid Selamariscina A against CYP2C8 and CYP2C9 Enzyme Activities in Human Liver Microsomes. Biflavonoids 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 32290339-0 2020 Strong and Selective Inhibitory Effects of the Biflavonoid Selamariscina A against CYP2C8 and CYP2C9 Enzyme Activities in Human Liver Microsomes. selamariscina a 59-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 32290339-7 2020 Returning to the P450 activities, selamariscina A inhibited CYP2C9-mediated diclofenac hydroxylation and tolbutamide hydroxylation with respective Ki values of 0.032 and 0.065 muM in a competitive and noncompetitive manner. Diclofenac 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 32290339-10 2020 This information might be useful in predicting herb-drug interaction potential between biflavonoids and co-administered drugs mainly metabolized by CYP2C8 and CYP2C9. Biflavonoids 87-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 32272615-0 2020 Lack of Correlation between In Vitro and In Vivo Studies on the Inhibitory Effects of (-)-Sophoranone on CYP2C9 is Attributable to Low Oral Absorption and Extensive Plasma Protein Binding of (-)-Sophoranone. sophoranone 88-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 32272615-3 2020 Of the nine tested CYPs, it exerted the strongest inhibitory effect on CYP2C9-mediated tolbutamide 4-hydroxylation with the lowest IC50 (Ki) value of 0.966 +- 0.149 muM (0.503 +- 0.0383 muM), in a competitive manner. Tolbutamide 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 32272615-4 2020 Additionally, it strongly inhibited other CYP2C9-catalyzed diclofenac 4"-hydroxylation and losartan oxidation activities. Diclofenac 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 32272615-4 2020 Additionally, it strongly inhibited other CYP2C9-catalyzed diclofenac 4"-hydroxylation and losartan oxidation activities. Losartan 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 32802593-3 2020 Fluvoxamine inhibits many cytochrome P450 enzyme (CYP) including CYP2C9, which metabolizes azilsartan. Fluvoxamine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 32802593-3 2020 Fluvoxamine inhibits many cytochrome P450 enzyme (CYP) including CYP2C9, which metabolizes azilsartan. azilsartan 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 32219822-11 2020 CONCLUSION: The polymorphisms of CYP2C9, VKORC1 and CYP4F2 genes can influence the stable dose of warfarin in Shandong population. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 32094096-7 2020 Interestingly, CYP2C9, CYP2C19, and CYP3A5 extensive metabolizers tended to be associated with lower cyclosporine C/D ratio when combined with voriconazole, but the results were not statistically significant. cyclosporine c 101-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 32094096-7 2020 Interestingly, CYP2C9, CYP2C19, and CYP3A5 extensive metabolizers tended to be associated with lower cyclosporine C/D ratio when combined with voriconazole, but the results were not statistically significant. Voriconazole 143-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 31673144-7 2020 Patients with CYP2C9 *1/*2, *1/*3, *2/*2, *2/*3, or *3/*3 required a lower warfarin maintenance dose compared with patients with CYP2C9 *1/*1 (SMD = -0.610, 95% CI: -0.802 to -0.419, I2 = 0%). Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 31653973-1 2020 The objective of this study is to estimate the prevalence of VKORC1, CYP2C9, and CYP4F2 genetic variants and their contribution to warfarin dose variability in Qataris. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 31653973-6 2020 Carriers of at least one CYP2C9 decreased function allele (*2 or *3) required lower median (IQR) warfarin doses compared to noncarriers [24.5 (14.5) mg/week vs. 35 (21) mg/week, p < 0.001]. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 31653973-11 2020 This study concluded that VKORC1 (-1639G>A), CYP2C9*2 & *3 are the most significant predictors of warfarin dose along with HTN, HF and smoking. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 31673144-11 2020 Our meta-analysis provides evidence that CYP2C9 and VKORC1 variant statuses affect warfarin maintenance dose in children, but not CYP4F2. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 32155912-5 2020 Therefore, in this in vitro study, we investigated the interactions of 24 microbial flavonoid metabolites with human serum albumin and cytochrome P450 (CYP2C9, 2C19, and 3A4) enzymes. Flavonoids 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 32155912-8 2020 All CYP isoforms examined were significantly inhibited by O-desmethylangolensin; nevertheless, only its effect on CYP2C9 seems to be relevant. O-desmethylangolensin 58-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 32071341-0 2020 Genome-Wide Association Study of VKORC1 and CYP2C9 on acenocoumarol dose, stroke recurrence and intracranial haemorrhage in Spain. Acenocoumarol 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 32106262-7 2020 During maintenance of human iPS-HLCs, ALB and urea producing abilities and CYP2C9, CYP2C19, and CYP3A4 activities were the highest when using HCM. hcm 142-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 32106262-11 2020 However, the CYP2C9, CYP2C19, and CYP3A4 activities were the highest when using HCM and WEM. hcm 80-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 32071341-9 2020 In conclusion, variants in VKORC1 and CYP2C9 are associated with acenocoumarol maintenance dose, stroke recurrence and ICH in a Spanish cohort. Acenocoumarol 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 31591509-6 2020 Most studies (78%) examined warfarin-CYP2C9/VKORC1 or clopidogrel-CYP2C19. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 31591509-11 2020 Supportive evidence exists for clopidogrel-CYP2C19 and warfarin-CYP2C9/VKORC1, but evidence is limited in other drug-gene combinations. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 32050158-5 2020 Therefore, the aim of this study was to develop, validate and optimize a simplified assay for the probe drugs caffeine (metabolized by CYP1A2), omeprazole (CYP2C19), losartan (CYP2C9), dextromethorphan (CYP2D6), midazolam (CYP3A4) and their respective enzyme-specific metabolites in small volumes (100 muL) of human plasma, that addresses the issues noted. Caffeine 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 32050158-5 2020 Therefore, the aim of this study was to develop, validate and optimize a simplified assay for the probe drugs caffeine (metabolized by CYP1A2), omeprazole (CYP2C19), losartan (CYP2C9), dextromethorphan (CYP2D6), midazolam (CYP3A4) and their respective enzyme-specific metabolites in small volumes (100 muL) of human plasma, that addresses the issues noted. Dextromethorphan 185-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 32050158-5 2020 Therefore, the aim of this study was to develop, validate and optimize a simplified assay for the probe drugs caffeine (metabolized by CYP1A2), omeprazole (CYP2C19), losartan (CYP2C9), dextromethorphan (CYP2D6), midazolam (CYP3A4) and their respective enzyme-specific metabolites in small volumes (100 muL) of human plasma, that addresses the issues noted. Midazolam 212-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 32071341-2 2020 Variants in CYP2C9 and VKORC1 have been associated with acenocoumarol maintenance dose. Acenocoumarol 56-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 32071341-3 2020 We analysed whether any of the 49 polymorphisms in CYP2C9 and VKORC1 previously associated with acenocoumarol maintenance dose in a Genome-Wide Association study (GWAs) in Dutch population are associated with stroke recurrence, intracranial haemorrhage (ICH) and acenocoumarol maintenance dose in a Spanish population. Acenocoumarol 96-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 31854189-0 2020 Different roles of human cytochrome P450 2C9 and 3A enzymes in diclofenac 4"- and 5-hydroxylations mediated by metabolically inactivated human hepatocytes in previously transplanted chimeric mice. Diclofenac 63-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-44 31875297-0 2020 Author"s Response to: "Letter to the Editor in Response to: "Effect of Polymorphisms in CYP2C9 and CYP2C19 on the Disposition, Safety and Metabolism of Progesterone Administrated Orally or Vaginally"". Progesterone 152-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 31973625-1 2020 The role of mirSNPs in the 3"UTR of VKORC1, CYP2C9 and CYP4F2 genes that could influence warfarin dose variability via a discrete miRNA-mediated mechanism remains unexplained. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 33758819-3 2020 Warfarin sodium is an oral anticoagulant that is primarily metabolized by cytochrome P450 2C9 (CYP2C9). Warfarin 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-93 33758819-3 2020 Warfarin sodium is an oral anticoagulant that is primarily metabolized by cytochrome P450 2C9 (CYP2C9). Warfarin 0-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 31744669-2 2020 A sensitive, specific, and fast ultra-high performance liquid chromatography/tandem mass spectrometry (UHPLC-MS/MS) method was developed and validated for determination of caffeine (probe of CYP1A2), tolbutamide (probe of CYP2C9), dextromethorphan (probe of CYP2D6), and alprazolam (probe of CYP3A4/5) in human serum. Caffeine 172-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 31875298-0 2020 Letter to the Editor in Response to "Effect of Polymorphisms in CYP2C9 and CYP2C19 on the Disposition, Safety and Metabolism of Progesterone Administrated Orally or Vaginally". Progesterone 128-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 31785970-0 2020 Cytochrome P450 2C9 polymorphism: Effect of amino acid substitutions on protein flexibility in the presence of tamoxifen. Tamoxifen 111-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 31657866-6 2020 RESULTS: Experiments with human liver microsomes and primary human hepatocytes in 3D co-culture confirmed that flurbiprofen is a suitable CYP2C9 substrate. Flurbiprofen 111-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-46 31679131-6 2020 Etravirine exhibits the potential for bi-directional drug-drug interactions with other antiretrovirals and concomitant medications through its interactions with cytochrome P450 (CYP) isozymes: CYP3A4, CYP2C9, and CYP2C19. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. afimoxifene 141-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-46 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. afimoxifene 141-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. Tamoxifen 150-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-46 31785970-1 2020 Tamoxifen is a prodrug and cytochrome P450 2C9 (CYP2C9) has a significant role in the formation of a therapeutically more potent metabolite (4-hydroxytamoxifen) than tamoxifen. Tamoxifen 150-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 31785970-5 2020 Therefore, in order to understand the genotype-phenotype association, it is aimed to study the interatomic interactions of amino acid substitutions in CYP2C9 variants in the presence of tamoxifen. Tamoxifen 186-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 31785970-6 2020 Computational structural biology approach was adopted to study the effect of amino acid substitutions of polymorphic variants of CYP2C9 R144C (*2), I359 L (*3), D360E (*5), R150H (*8), R335W (*11) and L90 P (*13) on the flexibility of the enzyme in the presence of tamoxifen. Tamoxifen 265-274 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 31785970-10 2020 Further, the effect of amino acid substitutions CYP2C9 variants on the protein flexibility in the presence of tamoxifen in 4-hydroxy orientation was evaluated by molecular dynamics (MD) simulations. Tamoxifen 110-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 31785970-14 2020 Thus, the interatomic interaction study of CYP2C9 variants in the presence of tamoxifen predicts the plausible effect of the investigated variants on the therapeutic outcome of tamoxifen. Tamoxifen 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 31785970-14 2020 Thus, the interatomic interaction study of CYP2C9 variants in the presence of tamoxifen predicts the plausible effect of the investigated variants on the therapeutic outcome of tamoxifen. Tamoxifen 177-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 31673875-12 2020 CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. Itraconazole 47-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 281-287 31673875-12 2020 CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. erdafitinib 138-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 31673875-1 2020 BACKGROUND AND OBJECTIVES: Erdafitinib, an oral selective pan-fibroblast growth factor receptor (FGFR) kinase inhibitor, is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. erdafitinib 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-174 31673875-2 2020 The aim of this phase 1 study was to assess the pharmacokinetics and safety of erdafitinib in healthy participants when coadministered with fluconazole (moderate CYP2C9 and CYP3A inhibitor), and itraconazole (a strong CYP3A4 and P-glycoprotein inhibitor). erdafitinib 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 31673875-12 2020 CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. erdafitinib 138-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 281-287 31673875-12 2020 CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. Fluconazole 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 281-287 31673875-12 2020 CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. erdafitinib 189-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 31720756-0 2020 Non-genetic factors and polymorphisms in genes CYP2C9 and VKORC1: predictive algorithms for TTR in Brazilian patients on warfarin. Warfarin 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 32047440-0 2019 Phenprocoumon Dose Requirements, Dose Stability and Time in Therapeutic Range in Elderly Patients With CYP2C9 and VKORC1 Polymorphisms. Phenprocoumon 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 31395958-0 2020 Algorithm for predicting low maintenance doses of warfarin using age and polymorphisms in genes CYP2C9 and VKORC1 in Brazilian subjects. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 32024122-6 2020 Reactions selective for flavin-containing monooxygenases (FMOs), CYP1B1, CYP2C9, CYP2J2, and CYP3A4 all show significant rates in human lung microsomal incubations, but all activities are higher when rat lung microsomes are used. Flavins 24-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 32433020-11 2020 Distinct expression of UGT1A1, CYP2B6, CYP2C9, CYP3A4, and CYP7A1 genes explains the ability to clear toxins and bilirubin as observed in normal hepatocytes. Bilirubin 113-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 32047440-1 2019 Background: Dose requirements of vitamin K antagonists are associated with CYP2C9 and VKORC1, but, compared to warfarin, less data is available about phenprocoumon. Vitamin K 33-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 32047440-3 2019 Methods: Aim was to scrutinize phenprocoumon dose requirements, dose stability and anticoagulation quality in association to CYP2C9 and VKORC1 in a natural cohort of elderly primary care patients. Phenprocoumon 31-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 31941150-2 2020 Herein, a new bergamottin containing a nitroxide moiety (SL-bergamottin, SL-BM, 10) was synthesized; chemically characterized, evaluated as a potential inhibitor of the CYP2C19, CYP3A4, and CYP2C9 enzymes; and compared to BM and known inhibitors such as ketoconazole (KET) (3A4), warfarin (WAR) (2C9), and ticlopidine (TIC) (2C19). bergamottin 14-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 31815452-5 2020 It was determined that UGT2B7 converts bromfenac to BI, and that while CYP2C8, CYP2C9 and CYP2C19 catalyze the hydroxylation of bromfenac, only CYP2C9 forms thioether adducts when incubated with NAC or GSH as trapping agents. bromfenac 128-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 31815452-5 2020 It was determined that UGT2B7 converts bromfenac to BI, and that while CYP2C8, CYP2C9 and CYP2C19 catalyze the hydroxylation of bromfenac, only CYP2C9 forms thioether adducts when incubated with NAC or GSH as trapping agents. Sulfides 157-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 31815452-5 2020 It was determined that UGT2B7 converts bromfenac to BI, and that while CYP2C8, CYP2C9 and CYP2C19 catalyze the hydroxylation of bromfenac, only CYP2C9 forms thioether adducts when incubated with NAC or GSH as trapping agents. Glutathione 202-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 32021384-0 2020 The Effect of UGT1A9, CYP2B6 and CYP2C9 Genes Polymorphism on Propofol Pharmacokinetics in Children. Propofol 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 32021384-1 2020 Purpose: This study was conducted to determine the effect of UGT1A9 98T>C, CYP2B6 516G>T and CYP2C9 430C>T genetic polymorphisms on the pharmacokinetics of propofol in children of different sexes and ages who undergone total intravenous anesthesia (TIVA) and deep sedation during diagnostic and therapeutic procedures. Propofol 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 32021384-9 2020 Conclusion: Further investigations of UGT1A9, CYP2B6 and CYP2C9 and other genes that participate in propofol metabolism as well as detailed analyses of the general conditions, administered therapies and associated diseases could explain the large interindividual variability of propofol metabolism in children. Propofol 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 32038232-1 2019 The cytochrome P450 2C9 and vitamin K epoxide reductase complex subunit 1 genotypes are associated with anticoagulation control and the clinical events in warfarin therapy. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-23 31941150-2 2020 Herein, a new bergamottin containing a nitroxide moiety (SL-bergamottin, SL-BM, 10) was synthesized; chemically characterized, evaluated as a potential inhibitor of the CYP2C19, CYP3A4, and CYP2C9 enzymes; and compared to BM and known inhibitors such as ketoconazole (KET) (3A4), warfarin (WAR) (2C9), and ticlopidine (TIC) (2C19). sl-bergamottin 57-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 32407272-1 2020 BACKGROUND: Metoclopramide is metabolized by various cytochrome P450 (CYP) enzymes such as CYP3A4, CYP1A2, CYP2D6, CYP2C9, and CYP2C19. Metoclopramide 12-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 31854268-4 2020 Conclusion: VKORC1 rs9923231, CYP2C9*3 rs1057910, ApoE rs7412, age, BMI and concomitant amiodarone play an important role in stable dose variation of warfarin in elderly Shanghai Han Chinese patients, whereas ABCB1 rs1045642 is not a significant genetic factor. Amiodarone 88-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 32228310-6 2020 CYP2C9*3 polymorphisms were associated with time to therapeutic range for acenocoumarol in Chilean patients, and the CYP4F2 TT genotype, MDR1 A allele, CYP1A2 A allele, and CYP3A4T allele are promising variants that merit further analysis. Acenocoumarol 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31378969-6 2020 Upadacitinib 30 mg once daily (a dose that is twice the optimal dose in rheumatoid arthritis based on phase 3 results) has a limited effect on CYP3A activity (26% decrease in exposure of midazolam, a sensitive CYP3A substrate) and no relevant effects on CYP1A2, CYP2C9, CYP2C19, or CYP2D6 activity in vivo. upadacitinib 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 262-268 31648049-11 2020 CYP2C9 and CYP2C19 were the two CYP isozymes that produced 3"-hydroxylnalbuphine and 4"-hydroxylnalbuphine. AN 3 59-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31648049-11 2020 CYP2C9 and CYP2C19 were the two CYP isozymes that produced 3"-hydroxylnalbuphine and 4"-hydroxylnalbuphine. BE 4-4-4-4 85-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31889870-1 2020 Warfarin doses are greatly affected by polymorphism altering cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) gene. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-80 33132262-2 2020 We developed a novel hypouricemic agent because benzbromarone, a commercially available uricosuric agent, has several problems, such as drug-induced liver injury or drug-drug interaction caused by CYP2C9 inhibition. Benzbromarone 48-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 31889870-1 2020 Warfarin doses are greatly affected by polymorphism altering cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) gene. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 31889870-11 2020 Both genotypes (CYP2C9 and VKORC1) require different dosing of warfarin than non-carriers in order to minimize the risk of warfarin overdosing and avoidance of the drug-related problems (DRPs). Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 31889870-11 2020 Both genotypes (CYP2C9 and VKORC1) require different dosing of warfarin than non-carriers in order to minimize the risk of warfarin overdosing and avoidance of the drug-related problems (DRPs). Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 31820397-1 2019 Zolpidem is extensively metabolized by CYP3A4, CYP2C9 and CYP1A2. zolpidem 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 31835701-0 2019 Influence of Selected Carbon Nanostructures on the CYP2C9 Enzyme of the P450 Cytochrome. Carbon 22-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 31835701-5 2019 The objective of this study was to evaluate the influence of three carbon nanostructures on the activity and expression at the mRNA and protein levels of CYP2C9 isoenzyme from the CYP2C subfamily: Diamond nanoparticles, graphite nanoparticles, and graphene oxide platelets. Carbon 67-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31835701-5 2019 The objective of this study was to evaluate the influence of three carbon nanostructures on the activity and expression at the mRNA and protein levels of CYP2C9 isoenzyme from the CYP2C subfamily: Diamond nanoparticles, graphite nanoparticles, and graphene oxide platelets. Diamond 197-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31835701-5 2019 The objective of this study was to evaluate the influence of three carbon nanostructures on the activity and expression at the mRNA and protein levels of CYP2C9 isoenzyme from the CYP2C subfamily: Diamond nanoparticles, graphite nanoparticles, and graphene oxide platelets. Graphite 220-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31835701-5 2019 The objective of this study was to evaluate the influence of three carbon nanostructures on the activity and expression at the mRNA and protein levels of CYP2C9 isoenzyme from the CYP2C subfamily: Diamond nanoparticles, graphite nanoparticles, and graphene oxide platelets. Graphite 248-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31835701-7 2019 A microsome model was used to assess the influence of the three-carbon nanostructures on the activity of the CYP2C9 isoenzyme. Carbon 64-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 32186616-2 2019 Its dosage is related to the genetic characteristics of CYP2C9 enzymatic activity, which metabolizes losartan to its active form E-3174, responsible for the antihypertensive effect. Losartan 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 31605674-6 2019 The findings confirmed that CYP3A4 was a major contributor (at least 30% total metabolism) to all three of the possible N-dealkylation pathways; however, CYP2C9, and not CYP2C19, played a critical role in terbinafine metabolism and even exceeded CYP3A4 contributions for terbinafine N-demethylation. Nitrogen 120-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31605674-6 2019 The findings confirmed that CYP3A4 was a major contributor (at least 30% total metabolism) to all three of the possible N-dealkylation pathways; however, CYP2C9, and not CYP2C19, played a critical role in terbinafine metabolism and even exceeded CYP3A4 contributions for terbinafine N-demethylation. terbinafine 205-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31605674-6 2019 The findings confirmed that CYP3A4 was a major contributor (at least 30% total metabolism) to all three of the possible N-dealkylation pathways; however, CYP2C9, and not CYP2C19, played a critical role in terbinafine metabolism and even exceeded CYP3A4 contributions for terbinafine N-demethylation. terbinafine 271-282 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31605674-6 2019 The findings confirmed that CYP3A4 was a major contributor (at least 30% total metabolism) to all three of the possible N-dealkylation pathways; however, CYP2C9, and not CYP2C19, played a critical role in terbinafine metabolism and even exceeded CYP3A4 contributions for terbinafine N-demethylation. Nitrogen 283-284 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31605674-11 2019 Taken together, the significance of CYP2C9 and 3A4 and to a lesser extent, CYP2C19, in terbinafine metabolism is consistent with reported drug interactions. terbinafine 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 31468576-0 2019 Drug interactions between warfarin and lenvatinib in a patient with the CYP2C9*1/*3 and VKORC1-1639G/A genotype. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 31468576-0 2019 Drug interactions between warfarin and lenvatinib in a patient with the CYP2C9*1/*3 and VKORC1-1639G/A genotype. lenvatinib 39-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 31468576-2 2019 (S)-Warfarin is metabolized to (S)-7-hydroxywarfarin by CYP2C9 and (S)-4"-hydroxywarfarin by CYP2C8. Warfarin 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 31468576-2 2019 (S)-Warfarin is metabolized to (S)-7-hydroxywarfarin by CYP2C9 and (S)-4"-hydroxywarfarin by CYP2C8. 7-hydroxywarfarin 31-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 31468576-3 2019 Here, we report drug interactions between warfarin and lenvatinib in a patient with CYP2C9*1/*3. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 31468576-3 2019 Here, we report drug interactions between warfarin and lenvatinib in a patient with CYP2C9*1/*3. lenvatinib 55-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 31468576-8 2019 WHAT IS NEW AND CONCLUSION: Lenvatinib-dependent (S)-warfarin inhibition could involve CYP2C9 and CYP2C8. lenvatinib 28-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 31468576-8 2019 WHAT IS NEW AND CONCLUSION: Lenvatinib-dependent (S)-warfarin inhibition could involve CYP2C9 and CYP2C8. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 31274208-0 2019 Prolonged Pharmacokinetic Interaction Between Capecitabine and a CYP2C9 Substrate, Celecoxib. Capecitabine 46-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 31274208-0 2019 Prolonged Pharmacokinetic Interaction Between Capecitabine and a CYP2C9 Substrate, Celecoxib. Celecoxib 83-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 31274208-1 2019 This study investigated the time course and magnitude of the pharmacokinetic interaction between capecitabine and the cytochrome P450 (CYP) 2C9 substrate celecoxib, with implications for coadministration of fluoropyrimidines with CYP2C9 substrates such as warfarin. Capecitabine 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-143 31274208-1 2019 This study investigated the time course and magnitude of the pharmacokinetic interaction between capecitabine and the cytochrome P450 (CYP) 2C9 substrate celecoxib, with implications for coadministration of fluoropyrimidines with CYP2C9 substrates such as warfarin. Capecitabine 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 31274208-1 2019 This study investigated the time course and magnitude of the pharmacokinetic interaction between capecitabine and the cytochrome P450 (CYP) 2C9 substrate celecoxib, with implications for coadministration of fluoropyrimidines with CYP2C9 substrates such as warfarin. Celecoxib 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-143 31274208-1 2019 This study investigated the time course and magnitude of the pharmacokinetic interaction between capecitabine and the cytochrome P450 (CYP) 2C9 substrate celecoxib, with implications for coadministration of fluoropyrimidines with CYP2C9 substrates such as warfarin. Celecoxib 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 230-236 31274208-1 2019 This study investigated the time course and magnitude of the pharmacokinetic interaction between capecitabine and the cytochrome P450 (CYP) 2C9 substrate celecoxib, with implications for coadministration of fluoropyrimidines with CYP2C9 substrates such as warfarin. Warfarin 256-264 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-143 31274208-7 2019 Close monitoring should be undertaken when administering fluoropyrimidines with CYP2C9 substrates with narrow therapeutic indexes while also weighing the benefits and risks for individual patients. fluoropyrimidines 57-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 32186616-2 2019 Its dosage is related to the genetic characteristics of CYP2C9 enzymatic activity, which metabolizes losartan to its active form E-3174, responsible for the antihypertensive effect. losartan carboxylic acid 129-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 32186616-10 2019 A non-significant tendency to need a larger dose of losartan was observed with the CYP2C9 * 3 allele, with an odds ratio (OR) of 1.46 (95% confidence intervals (CI) 0.01-18.64). Losartan 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 32186616-13 2019 The presence of CYP2C9*3 is associated with the need for higher doses of losartan, possibly due to a decrease in the conversion of losartan to E-3174. Losartan 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32186616-13 2019 The presence of CYP2C9*3 is associated with the need for higher doses of losartan, possibly due to a decrease in the conversion of losartan to E-3174. Losartan 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 32186616-13 2019 The presence of CYP2C9*3 is associated with the need for higher doses of losartan, possibly due to a decrease in the conversion of losartan to E-3174. losartan carboxylic acid 143-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 31420941-2 2019 We previously constructed a physiologically-based pharmacokinetic (PBPK) model of rifampicin accounting for the components for the induction of cytochrome P450 (CYP) 3A/CYP2C9 and the inhibition of organic anion transporting polypeptide 1B (OATP1B). Rifampin 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 31199498-1 2019 We predicted the drug-drug interaction (DDI) potential of siponimod in presence of cytochrome P450 (CYP)2C9/CYP3A4 inhibitors/inducers in subjects with different CYP2C9 genotypes by physiologically-based pharmacokinetic (PK) modeling. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-107 31199498-1 2019 We predicted the drug-drug interaction (DDI) potential of siponimod in presence of cytochrome P450 (CYP)2C9/CYP3A4 inhibitors/inducers in subjects with different CYP2C9 genotypes by physiologically-based pharmacokinetic (PK) modeling. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 31392364-0 2019 Siponimod pharmacokinetics, safety, and tolerability in combination with the potent CYP3A4 inhibitor itraconazole in healthy subjects with different CYP2C9 genotypes. siponimod 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 31318064-7 2019 These interesting results steered to enumerate the possible role of IS01957 towards diclofenac pharmacokinetics through a panel of mechanistic investigations: (a) BCRP dependent ATPase activity was markedly interfered by IS01957; (b) IS01957 increased the intestinal permeability of diclofenac in the single pass in-situ perfusion model; (c) IS01957 inhibited the CYP2C9 catalyzed diclofenac 4-hydroxylation in human liver microsomes. is01957 68-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 364-370 31318064-7 2019 These interesting results steered to enumerate the possible role of IS01957 towards diclofenac pharmacokinetics through a panel of mechanistic investigations: (a) BCRP dependent ATPase activity was markedly interfered by IS01957; (b) IS01957 increased the intestinal permeability of diclofenac in the single pass in-situ perfusion model; (c) IS01957 inhibited the CYP2C9 catalyzed diclofenac 4-hydroxylation in human liver microsomes. Diclofenac 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 364-370 31392364-1 2019 PURPOSE: To evaluate the PK and safety of siponimod, a substrate of CYP2C9/3A4, in the presence or absence of a CYP3A4 inhibitor, itraconazole. siponimod 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 31640208-7 2019 About 30 genes implicated in the metabolism coumarins derivatives were identified, the most important being CYP2C9 and VKORC, each with several polymorphisms. coumarin 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 31064211-7 2019 In patients with wild-type genotypes for VKORC1, CYP2C9*2, CYP2C9*3, and CYP4F2, the highest warfarin requirement was found in Caucasians, lower in Hispanics, and lowest in Asians. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 31064211-7 2019 In patients with wild-type genotypes for VKORC1, CYP2C9*2, CYP2C9*3, and CYP4F2, the highest warfarin requirement was found in Caucasians, lower in Hispanics, and lowest in Asians. Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 31064211-10 2019 CYP2C9 variants were associated with lower ADW doses; frequencies of CYP2C9*2 and CYP2C9*3 mutations were higher in Caucasians than in Hispanics but rare to none in Asians. ADW 742 43-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31676845-7 2019 We also demonstrated that matured Huh7s can be used for drug induction studies, where CYP3A4, CYP1A2, CYP2C9, and CYP2C19 inductions were achieved following rifampicin treatment. Rifampin 157-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 31731555-4 2019 Furthermore, the inhibitory effects of DHF on cytochrome P450 (CYP2C9, CYP2C19, and CYP3A4) and xanthine oxidase (XO) enzymes were also tested using in vitro models. 6,7-dihydroxyflavone 39-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 31515991-3 2019 Auriculasin inhibited UGT1A6, UGT1A8, UGT1A10, UGT2B7, CYP2C9, and CYP3A4 strongly at a concentration of 100 muM. auriculasin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 31515991-6 2019 In addition, CYP2D6, CYP2C9, CYP2C19, and CYP2C8 were the major CYP isoforms involved in the metabolism of auriculasin. auriculasin 107-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 31276316-7 2019 Unexpectedly, several mutants showed significantly improved activity towards CYP2C9 (mutant 1-014) and/or CYP2A6 (mutants 1-014, 1-015, 1-053 and 1-077) using NADPH, even though the mutations were introduced at locations remote from the putative CPR-P450 interaction face. NADP 159-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 30557096-7 2019 The results indicated that catalpol could inhibit the activity of CYP3A4, CYP2E1 and CYP2C9, with IC50 values of 14.27, 22.4 and 14.69 muM, respectively, but those other CYP isoforms were not affected. catalpol 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 31616008-3 2019 Mechanistically, we also found a gender-specific correlation of the CYP2C9rs1799853T/T genotype with serum vitamin D3 levels (P = 0.00085) and a modest effect on IL1beta levels after stimulation of PBMCs or blood with LPS and PHA (P = 0.0057 and P = 0.0058). Cholecalciferol 107-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 31176252-9 2019 Furthermore, reaction phenotyping studies revealed that, despite the low hepatic clearance in the first pass metabolism of TEB, multiple human CYP450 isoforms were involved in TEB metabolism when TEBOH enantiomers were generated, mainly CYP3A4 and CYP2C9, which makes TEB accumulation in the human body more difficult due to multiple metabolic pathways. tebuconazole 123-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 248-254 31324935-1 2019 PURPOSE: Ruxolitinib is metabolized by cytochrome P450 (CYP)3A4 and CYP2C9. ruxolitinib 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 31482508-0 2019 Effect of Polymorphisms in CYP2C9 and CYP2C19 on the Disposition, Safety and Metabolism of Progesterone Administrated Orally or Vaginally. Progesterone 91-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 31482508-6 2019 CONCLUSION: This is the first study that reports variability in progesterone disposition according to the CYP2C19 and CYP2C9 phenotype. Progesterone 64-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 31114948-2 2019 Cytochrome P450 (CYP) 2C9, the primary enzyme responsible for THC metabolism, has two single nucleotide polymorphisms-Arg144Cys (*2) and Ile359Leu (*3). Dronabinol 62-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 31399508-8 2019 These results strongly support a high propensity for rhinacanthin-C as a perpetrator of clinical herb-drug interaction via inhibiting various influx and efflux drug transporters (i.e., P-gp, BCRP, OATP1B1, and OATP1B3) and CYP isoforms (i.e., CYP2C8, CYP2C9, and CYP2C19). rhinacanthin-C 53-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 251-257 31058419-2 2019 Amlodipine is mainly metabolized by cytochrome P450 (CYP) 3A4, whereas losartan is metabolized by CYP2C9 and CYP3A4. Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 30825074-4 2019 METHODS: This study was performed to investigate the inhibitory effects of myricetin on human CYP3A4, CYP2B6, CYP2C9, CYP2D6 and rat CYP3A2, CYP2B1, CYP2C11, CYP2D1 through the cocktail approach using ultra-performance liquid chromatography tandem mass spectrometry. myricetin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 31114948-6 2019 We therefore sought to characterize the pharmacokinetics of THC and its major metabolites 11-hydroxy-delta-9-tetrahydrocannabinol (THC-OH) and 11-nor-9-carboxy-delta-9-tetrahydrocannabinol (THC-COOH) in healthy volunteers with known CYP2C9 status by non-compartmental analysis (NCA), compartmental modeling (CM) and minimal physiologically based pharmacokinetic (mPBPK) modeling. Dronabinol 60-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 31114948-11 2019 THC hepatic clearance is dependent on the CYP2C9 genetic variant in the population. Dronabinol 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 31114948-14 2019 THC-OH is also a high extraction ratio drug, but its hepatic clearance is significantly impacted by the hepatic diffusional barrier that impedes its access to hepatic CYP2C9. thc-oh 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 31114948-15 2019 THC-COOH is glucuronidated and renally cleared; subjects homozygous for CYP2C9*3 have reduced exposure to this metabolite as a result of the polymorphism reducing THC production, the hepatic diffusional barrier impeding egress from the hepatocyte, and increased renal clearance. Dronabinol 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 31114948-15 2019 THC-COOH is glucuronidated and renally cleared; subjects homozygous for CYP2C9*3 have reduced exposure to this metabolite as a result of the polymorphism reducing THC production, the hepatic diffusional barrier impeding egress from the hepatocyte, and increased renal clearance. Carbonic Acid 4-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 31114948-15 2019 THC-COOH is glucuronidated and renally cleared; subjects homozygous for CYP2C9*3 have reduced exposure to this metabolite as a result of the polymorphism reducing THC production, the hepatic diffusional barrier impeding egress from the hepatocyte, and increased renal clearance. Dronabinol 163-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 31114948-16 2019 CONCLUSION: It has recently been reported that the terminal metabolite THC-COOH is active, implying the exposure difference in individuals homozygous for CYP2C9*3 may become therapeutically relevant. Dronabinol 71-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31114948-16 2019 CONCLUSION: It has recently been reported that the terminal metabolite THC-COOH is active, implying the exposure difference in individuals homozygous for CYP2C9*3 may become therapeutically relevant. Carbonic Acid 75-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31186542-5 2019 In addition to selected SNPs, VKORC1 g.6399C>T, and CYP2C9 c.1075A>C, which were known to have significant effects on warfarin stable doses, were also included in the study. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 31421085-6 2019 KEY FINDINGS: At 100 muM, licoricidin strongly inhibited CYP2C9, CYP2C19, CYP3A4, UGT1A3, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT2B4, UGT2B7, UGT2B15, and UGT2B17. licoricidin 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 33061049-2 2019 CYP2C9 is a hepatic drug-metabolizing enzyme in the CYP450 superfamily and is the primary metabolizing enzyme of warfarin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31461080-0 2019 Assessing the clinical impact of CYP2C9 pharmacogenetic variation on phenytoin prescribing practice and patient response in an integrated health system. Phenytoin 69-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 33061049-8 2019 Majority of the patients, the 83 in warfarin group and the 40 in acenocoumarol group, had a wild CYP2C9 diplotype. Acenocoumarol 65-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 29676195-8 2019 Enzyme kinetic studies showed that sophocarpine is not only a noncompetitive inhibitor of CYP3A4 but also a competitive inhibitor of CYP2C9, with Ki values of 6.74 and 9.19 muM, respectively. sophocarpine 35-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 31461080-1 2019 OBJECTIVE: To assess the impact of CYP2C9 variation on phenytoin patient response and clinician prescribing practice where genotype was unknown during treatment. Phenytoin 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 31461080-3 2019 We used laboratory test results, medication dispensing records, and medical notes to identify associations of CYP2C9 genotype with phenytoin blood concentration, neurologic side effects, and medication dispensing patterns reflecting clinician prescribing practice and patient response. Phenytoin 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 31461080-6 2019 Decreased function CYP2C9 genotypes were associated with medication dispensing patterns indicating dose decrease, use of alternative anticonvulsants, and worse adherence, although these associations varied by treatment indication for phenytoin. Phenytoin 234-243 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 31206740-5 2019 We also describe an additional case of phenytoin DRESS (RegiSCAR DRESS score 7) in an Aboriginal Australian that was associated with HLA-B*56:02 and with CYP2C9*1/*3 genotype. Phenytoin 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 31492893-5 2019 Based on combined polymorphisms in CYP2C9 and VKORC1, we established a clinical classification for warfarin sensitivity. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 31259734-0 2019 AKR1D1*36 C>T (rs1872930) allelic variant is associated with variability of the CYP2C9 genotype predicted pharmacokinetics of ibuprofen enantiomers - a pilot study in healthy volunteers. Ibuprofen 126-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 31259734-1 2019 The relative contribution of CYP2C9 allelic variants to the pharmacokinetics (PK) of ibuprofen (IBP) enantiomers has been studied extensively, but the potential clinical benefit of pharmacogenetically guided IBP treatment is not evident yet. Ibuprofen 85-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 31487853-5 2019 We describe herein the modeling and simulation of CYP 2C9 and CYP 2C19 in a phospholipid bilayer. Phospholipids 76-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-57 30989645-3 2019 CYP2C9*3 associated with significantly increased area under the plasma concentration-time curve (AUC) of both 3R,5S-fluvastatin and 3S,5R-fluvastatin (by 67% and 94% per variant allele copy, P = 3.77 x 10-9 and P = 3.19 x 10-12 ). 3r,5s-fluvastatin 110-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30989645-3 2019 CYP2C9*3 associated with significantly increased area under the plasma concentration-time curve (AUC) of both 3R,5S-fluvastatin and 3S,5R-fluvastatin (by 67% and 94% per variant allele copy, P = 3.77 x 10-9 and P = 3.19 x 10-12 ). 3s,5r-fluvastatin 132-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30989645-5 2019 A candidate gene analysis suggested that CYP2C9*2 also affects the AUC of both fluvastatin enantiomers and that SLCO2B1 single-nucleotide variations may affect the AUC of 3S,5R-fluvastatin. Fluvastatin 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 30989645-7 2019 Genotyping of both CYP2C9 and SLCO1B1 may be useful in predicting fluvastatin efficacy and myotoxicity. Fluvastatin 66-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 31336317-2 2019 GFZ may affect the metabolism of various drugs, including statins, by inhibiting the sinusoidal influx transporter OATP1B1 and also CYP2C9 and CYP2C8 enzymes. Gemfibrozil 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 31453773-0 2019 Low-frequency variants at the CYP2C9 locus among Puerto Rican patients on warfarin: in silico predictions of functionality and conservation. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 31075510-7 2019 A separate recommendation on warfarin PGx testing is being developed to include recommendations on CYP2C9 alleles and additional warfarin sensitivity-associated genes and alleles. Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 31447576-0 2019 Polymorphisms of CYP2C9*2, CYP2C9*3 and VKORC1 genes related to time in therapeutic range in patients with atrial fibrillation using warfarin. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 31447576-0 2019 Polymorphisms of CYP2C9*2, CYP2C9*3 and VKORC1 genes related to time in therapeutic range in patients with atrial fibrillation using warfarin. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 31447576-11 2019 Conclusion: The genotypes of the CYP2C9*3 (AA) and VKORC1 -1639 (GG) genes were associated with the worst quality of anticoagulation control (TTR) in patients with AF using warfarin in the northeast of Brazil. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Cannabinoids 91-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). delta-tetrahydrocannabinol 225-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Dronabinol 253-256 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Cannabidiol 259-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Cannabidiol 272-275 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Cannabinol 241-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-6 2019 FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Delta-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). Cannabinol 294-297 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 31433338-9 2019 Delta-Tetrahydrocannabinol also activates CYP2C9 and induces CYP1A1. delta-tetrahydrocannabinol 0-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 31163215-3 2019 All tested coumarins except a C4 phenyl derivative (11) showed higher inhibitory activities for CYP2A6 over the other CYP isoforms, including CYP1A2, CYP2D6, CYP2E1, CYP3A4, CYP2C8, and CYP2C9. Coumarins 11-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 186-192 31486736-0 2019 CYP2C9*2 is associated with indomethacin treatment failure for patent ductus arteriosus. Indomethacin 28-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31453773-3 2019 Results: CYP2C9*5,*8,*9,*11,*12,*21 and a novel *61 induce conformational changes that affect the binding site of S-warfarin. Warfarin 114-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 31486736-5 2019 Conclusion: Age, surfactant use, and CYP2C9*2 influence indomethacin treatment outcome in preterm infants with PDA. Indomethacin 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 31453773-5 2019 Conclusion: The unfavorable distance of S-warfarin from heme group, and low-binding interactions due to these CYP2C9 variants, suggest major complications during warfarin therapy. Warfarin 162-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 30675694-3 2019 METHODS: A PBPK model of hyperforin accounting for the induction of cytochrome P450 (CYP) 3A, CYP2C9 and CYP2C19 was developed in the Simcyp Simulator (version 17) and verified using published, clinically observed pharmacokinetic data. hyperforin 25-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 31068367-5 2019 To describe the pharmacokinetic interaction profile of alectinib in a complete fashion, its possible inhibitory properties toward clinically relevant P450 enzymes (i.e., CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP3A4, or CYP3A5) were evaluated using human P450-expressing insect microsomes, revealing alectinib as a poor interactor. alectinib 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 30852642-1 2019 PURPOSE: Large inter-individual differences in warfarin maintenance dose are mostly due to the effect of genetic polymorphisms in multiple genes, including vitamin K epoxide reductase complex 1 (VKORC1), cytochromes P450 2C9 (CYP2C9), and cytochrome P450 4F2 (CYP4F2). Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 204-224 30852642-1 2019 PURPOSE: Large inter-individual differences in warfarin maintenance dose are mostly due to the effect of genetic polymorphisms in multiple genes, including vitamin K epoxide reductase complex 1 (VKORC1), cytochromes P450 2C9 (CYP2C9), and cytochrome P450 4F2 (CYP4F2). Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 226-232 30675694-8 2019 CONCLUSION: In the current study, a PBPK model for hyperforin was successfully developed, with a predictive capability for the interactions of SJW with different CYP3A, CYP2C9 and CYP2C19 substrates. hyperforin 51-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 30758919-2 2019 In vitro, lesinurad was shown to be a weak inhibitor of cytochrome P450 (CYP)2C9 and a weak inducer of CYP3A4. lesinurad 10-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-80 31048453-7 2019 CYP2C9 pathway was the major pathway for depletion of THC (fm = 0.91, Km,u = 3 nM) and formation of 11-OH-THC. Dronabinol 54-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31048453-7 2019 CYP2C9 pathway was the major pathway for depletion of THC (fm = 0.91, Km,u = 3 nM) and formation of 11-OH-THC. 11-Hydroxytetrahydrocannabinol 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 31608877-0 2019 Relationship of S/R warfarin ratio with CYP2C9 genotypes in Pakistani population. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 31048453-9 2019 11-OH-THC was depleted by UGTs (fm = 0.67 and Km,u = 39 nM), CYP3A4 (fm = 0.18, Km,u = 824 nM), and CYP2C9 (fm = 0.15, Km,u = 33 nM). 11-Hydroxytetrahydrocannabinol 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 29945531-1 2019 BACKGROUND: Voriconazole is extensively metabolized by the CYP450 isoenzymes 2C19 and 3A4 and to a lesser extent by CYP2C9; therefore, any medication that affects this pathway can alter its plasma concentration. Voriconazole 12-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 31608877-3 2019 This study was carried out to determine the effect of CYP2C9*2 and CYP2C9*3 polymorphisms on S/R warfarin ratio. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 31608877-3 2019 This study was carried out to determine the effect of CYP2C9*2 and CYP2C9*3 polymorphisms on S/R warfarin ratio. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 31608877-7 2019 The concentration of S-warfarin was significantly different among CYP2C9 genotypes (p =0.018) whereas there was no effect on R-warfarin (p =0.134). Warfarin 21-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 31608877-8 2019 There was statistically significant effect of different CYP2C9 genotypes on S/R warfarin ratio (p=0.000). Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 31608877-9 2019 It is concluded that CYP2C9 polymorphisms influence CYP2C9 enzymatic activity in turn affecting S-warfari levels but not R-warfarin, thus leading to different S/R warfarin enantiomers ratio among different CYP2C9 genotypes. s-warfari 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 31608877-9 2019 It is concluded that CYP2C9 polymorphisms influence CYP2C9 enzymatic activity in turn affecting S-warfari levels but not R-warfarin, thus leading to different S/R warfarin enantiomers ratio among different CYP2C9 genotypes. s-warfari 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 31608877-9 2019 It is concluded that CYP2C9 polymorphisms influence CYP2C9 enzymatic activity in turn affecting S-warfari levels but not R-warfarin, thus leading to different S/R warfarin enantiomers ratio among different CYP2C9 genotypes. s-warfari 96-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 30506689-0 2019 Effect of CYP4F2, VKORC1, and CYP2C9 in Influencing Coumarin Dose: A Single-Patient Data Meta-Analysis in More Than 15,000 Individuals. coumarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 31239454-8 2019 Furthermore, the results indicate that coumarin (CYP2A), midazolam (CYP3A), tolbutamide (CYP2C), and dextromethorphan (CYP2D) are as selective for porcine as for human CYP450. Tolbutamide 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-94 30306496-0 2019 Effect of 95% Ethanol Khat Extract and Cathinone on in vitro Human Recombinant Cytochrome P450 (CYP) 2C9, CYP2D6, and CYP3A4 Activity. cathinone 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-104 30992242-2 2019 Activities of CYP2C9, CYP2C19, and CYP3A4 were determined by using the prototypical substrates tolbutamide, omeprazole and midazolam, respectively. Midazolam 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 30992242-2 2019 Activities of CYP2C9, CYP2C19, and CYP3A4 were determined by using the prototypical substrates tolbutamide, omeprazole and midazolam, respectively. Tolbutamide 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 30992242-2 2019 Activities of CYP2C9, CYP2C19, and CYP3A4 were determined by using the prototypical substrates tolbutamide, omeprazole and midazolam, respectively. Omeprazole 108-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 30306496-4 2019 This study aimed to reveal the inhibitory potencies of khat ethanol extract (KEE) and its major active ingredient (cathinone) on human cytochrome P450 (CYP) 2C9, CYP2D6, and CYP3A4 enzymes activities, which are collectively responsible for metabolizing 70-80% clinically used drugs. Ethanol 60-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-160 30306496-4 2019 This study aimed to reveal the inhibitory potencies of khat ethanol extract (KEE) and its major active ingredient (cathinone) on human cytochrome P450 (CYP) 2C9, CYP2D6, and CYP3A4 enzymes activities, which are collectively responsible for metabolizing 70-80% clinically used drugs. cathinone 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-160 30955332-4 2019 The MeOH extract exhibited significant inhibition of the major human CYP450 isozymes (CYP3A4, CYP1A2, CYP2D6, CYP2C9, and CYP2C19). Methanol 4-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 30991128-11 2019 Overall, the present investigation suggested that myclobutanil moderately inhibits CYP2D6 and CYP2C9 in vitro and strongly inhibits CYP3A and CYP2C19 in vitro. systhane 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 30525241-0 2019 Association between polymorphisms of VKORC1 and CYP2C9 genes with warfarin maintenance dose in a group of warfarin users in Birjand city, Iran. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 30525241-0 2019 Association between polymorphisms of VKORC1 and CYP2C9 genes with warfarin maintenance dose in a group of warfarin users in Birjand city, Iran. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 30525241-3 2019 The aim of the present study was to evaluate the frequency of CYP2C9 and VKORC1 gene polymorphisms and their association with warfarin maintenance dose in a sample of cardiovascular patients in Birjand, South-Khorasan province of Iran. Warfarin 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 31145690-0 2019 Drug-drug interaction (DDI) assessments of ruxolitinib, a dual substrate of CYP3A4 and CYP2C9, using a verified physiologically based pharmacokinetic (PBPK) model to support regulatory submissions. ruxolitinib 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 31145690-1 2019 Ruxolitinib is mainly metabolized by cytochrome P450 (CYP) enzymes CYP3A4 and CYP2C9 followed by minor contributions of other hepatic CYP enzymes in vitro. ruxolitinib 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 31145690-2 2019 A physiologically based pharmacokinetic (PBPK) model was established to evaluate the changes in the ruxolitinib systemic exposures with co-administration of CYP3A4 and CYP2C9 perpetrators. ruxolitinib 100-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 31145690-3 2019 The fractions metabolized in the liver via oxidation by CYP enzymes (fm,CYP3A4 = 0.75, fm,CYP2C9 = 0.19, and fm,CYPothers = 0.06) for an initial ruxolitinib model based on in vitro data were optimized (0.43, 0.56, and 0.01, respectively) using the observed exposure changes of ruxolitinib (10 mg) with co-administered ketoconazole (200 mg). ruxolitinib 145-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 31145690-9 2019 Furthermore, an AUC increase of ruxolitinib in the absence or presence of the dual CYP3A4 and CYP2C9 inhibitor fluconazole (100-400 mg) was prospectively estimated to be 1.94- to 4.31-fold. ruxolitinib 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 31145690-9 2019 Furthermore, an AUC increase of ruxolitinib in the absence or presence of the dual CYP3A4 and CYP2C9 inhibitor fluconazole (100-400 mg) was prospectively estimated to be 1.94- to 4.31-fold. Fluconazole 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 31145690-11 2019 A ruxolitinib PBPK model with optimized fm,CYP3A4 and fm,CYP2C9 was established to evaluate victim DDI risks. ruxolitinib 2-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 31145690-12 2019 The previous minimal PBPK model was supported by the FDA for the dose reduction strategy, halving the dose with the concomitant use of strong CYP3A4 inhibitors and dual inhibitors on CYP2C9 and CYP3A4, such as fluconazole at <=200 mg. Fluconazole simulation results were used as supportive evidence in discussions with the FDA and EMA about ruxolitinib dose adjustment when co-administering perpetrator drugs. Fluconazole 210-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 31086662-0 2019 Frequency of CYP2C9 (*2, *3 and IVS8-109A>T) allelic variants, and their clinical implications, among Mexican patients with diabetes mellitus type 2 undergoing treatment with glibenclamide and metformin. Glyburide 178-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 31086662-0 2019 Frequency of CYP2C9 (*2, *3 and IVS8-109A>T) allelic variants, and their clinical implications, among Mexican patients with diabetes mellitus type 2 undergoing treatment with glibenclamide and metformin. Metformin 196-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 31086662-1 2019 The majority of Mexican patients with diabetes mellitus type 2 (DMT2) (67.9-85.0%) are prescribed sulphonylureas (SUs), which are metabolized by cytochrome P450 2C9 (abbreviated as CYP2C9). Sulfonylurea Compounds 114-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-164 31086662-1 2019 The majority of Mexican patients with diabetes mellitus type 2 (DMT2) (67.9-85.0%) are prescribed sulphonylureas (SUs), which are metabolized by cytochrome P450 2C9 (abbreviated as CYP2C9). Sulfonylurea Compounds 114-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 30730615-6 2019 Overall, ASP8477 was a weak inhibitor for CYP3A4 and CYP2C9, a moderate to strong inhibitor for CYP2C19, and a weak to strong inhibitor for CYP2D6, with doses from 20 to 100 mg. pyridin-3-yl 4-(phenylcarbamoyl)piperidine-1-carboxylate 9-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 30821933-0 2019 VKORC1 and Novel CYP2C9 Variation Predict Warfarin Response in Alaska Native and American Indian People. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 31037455-8 2019 CYP2C9 *1/*3, VKORC1 AA, concomitant amiodarone, and nonheart valve replacement reduced the warfarin Km by 30.4%, 74.3%, 34.5%, and 39.4%, respectively. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30770140-1 2019 BACKGROUND: Amiodarone, which inhibits CYP2C9 and P-glycoprotein, is commonly prescribed with non-vitamin K antagonist oral anticoagulants (NOACs) and polypharmacy in high-risk atrial fibrillation (AF) patients. Amiodarone 12-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 30589990-2 2019 Warfarin was the most extensively studied drug in a PGx context: a genomewide association study targeting warfarin stable dose identified significant signals in VKORC1 and CYP2C9, several PGx dosing algorithms were developed based on these and other genes, and the implications of population admixture on extrapolation of dosing recommendations in the CPIC guidelines were examined. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 30589990-2 2019 Warfarin was the most extensively studied drug in a PGx context: a genomewide association study targeting warfarin stable dose identified significant signals in VKORC1 and CYP2C9, several PGx dosing algorithms were developed based on these and other genes, and the implications of population admixture on extrapolation of dosing recommendations in the CPIC guidelines were examined. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 29779438-3 2019 Other CYP enzymes contributing to the metabolism of one or more of the three pyrethroids were CYP1A2, CYP2C8, CYP2C9*1, CYP2D6*1, CYP3A4 and CYP3A5. Pyrethrins 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 31061616-0 2019 Effects of CYP2C9 and VKORC1 polymorphisms on warfarin sensitivity and responsiveness during the stabilization phase of therapy. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 31061616-1 2019 The main objective of this study is to assess the effects of CYP2C9 and VKORC1 polymorphisms on warfarin sensitivity and responsiveness in a Jordanian population during the stabilization phase of treatment. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 31061616-6 2019 Carriers of the CYP2C9 polymorphisms rs1057910 and rs4086116 had an increased risk of warfarin sensitivity compared to subjects with no or only one polymorphism. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 31061616-9 2019 The presence of any of CYP2C9 or VKORC1 polymorphisms is associated with sensitivity to warfarin during the stabilization period. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 30903874-9 2019 Furthermore, single nucleotide polymorphisms (SNPs) in CYP2C9, VKORC1, and MIR133 genes can help physicians to predict individual warfarin dose requirement. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 31531081-7 2019 Statistically significant association between clinically optimized warfarin dose and indication for the treatment, age, and warfarin sensitivity determined by VKORC1, CYP2C9 gene polymorphisms were confirmed. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 30520061-8 2019 The LC-MS/MS data showed DHI intensively inhibit CYP2A6 activity and the intensity of inhibition was followed by CYP2C8, CYP3A4, CYP2C19, CYP2B6, CYP2D6, CYP1A2, CYP2E1 and CYP2C9 in vitro. dehydrosoyasaponin I 25-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 31531081-7 2019 Statistically significant association between clinically optimized warfarin dose and indication for the treatment, age, and warfarin sensitivity determined by VKORC1, CYP2C9 gene polymorphisms were confirmed. Warfarin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 29298995-1 2019 Warfarin is primarily metabolized by cytochrome 2C9, encoded by gene CYP2C9. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 30983536-0 2019 Extremely low therapeutic doses of acenocoumarol in a patient with CYP2C9*3/*3 and VKORC1-1639A/A genotype. Acenocoumarol 35-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30983536-4 2019 We present the first reported case of a patient with the compounded genotype CYP2C9*3*3 and VKORC1-1639A/A under treatment with acenocoumarol, and review of other reported cases with analogous genotypic profiles but under treatment with warfarin. Acenocoumarol 128-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 30983536-4 2019 We present the first reported case of a patient with the compounded genotype CYP2C9*3*3 and VKORC1-1639A/A under treatment with acenocoumarol, and review of other reported cases with analogous genotypic profiles but under treatment with warfarin. Warfarin 237-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 30789308-0 2019 Polymorphisms in CYP1A2, CYP2C9 and ABCB1 affect agomelatine pharmacokinetics. agomelatine 49-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 30712247-0 2019 Interpretation of the effect of CYP2C9, VKORC1 and CYP4F2 variants on warfarin dosing adjustment in Turkey. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 30521883-4 2019 Increased expression of CYP2C epoxygenases, together with increased levels of their arachidonic acid-derived products, is often observed in tumors and tumor-associated vasculature, making these enzymes an ideal target for anti-cancer therapies. Arachidonic Acid 84-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-29 30562214-0 2019 Individuals with CYP2C8 and CYP2C9 reduced metabolism haplotypes self-adjusted ibuprofen dose in the Coriell Personalized Medicine Collaborative. Ibuprofen 79-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 30562214-1 2019 OBJECTIVES: The objectives of this study were to determine whether differences in CYP2C8 and CYP2C9 haplotype influence the dose of ibuprofen self-administered by individuals, and to examine the potential relationship between CYP2C8 and CYP2C9 reduced metabolism haplotypes and adverse events. Ibuprofen 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 30562214-2 2019 PARTICIPANTS AND METHODS: We investigated relationships between genetic variations in CYP2C8 and CYP2C9 and ibuprofen use, dose, and side effects (reported by questionnaire) in 445 participants from the Coriell Personalized Medicine Collaborative. Ibuprofen 108-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 30562214-3 2019 RESULTS: Carriers of reduced metabolism haplotypes for CYP2C8 (*2, *3, *4) and CYP2C9 (*2, *3) were significantly (P=0.0171) more likely than those lacking these variants to take less than the recommended dose of ibuprofen, after controlling for sex, age, race, and cohort. Ibuprofen 213-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 30562214-6 2019 CONCLUSION: These results suggest a subset of individuals with genetic variation in CYP2C8 and CYP2C9 recognize that they obtain adequate drug efficacy with lower ibuprofen doses, or take lower doses due to prior side effects. Ibuprofen 163-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 29298995-2 2019 Here, we investigated whether variants in nuclear receptor genes which regulate the expression of CYP2C9 are associated with warfarin response. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 30962704-0 2019 Impact of a variable number tandem repeat in the CYP2C9 promoter on warfarin sensitivity and responsiveness in Jordanians with cardiovascular disease. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 30962704-1 2019 Purpose: The purpose of this study was to investigate the influence of CYP/CYP450 2C9 (CYP2C9) promoter variable number tandem repeat (p-VNTR) polymorphism on susceptibility to cardiovascular disease and on warfarin sensitivity and responsiveness, in Jordanians with cardiovascular disease during initiation and stabilization phases of therapy. Warfarin 207-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 30962704-3 2019 PCR-based methods were performed to analyze the effects of CYP2C9 p-VNTR polymorphism on warfarin metabolism. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 30690186-9 2019 Atipamezole has more effective inhibition to CYP2C9 mediated diclofenac hydroxylation in human and animal liver microsomes with IC50 values of 1.50-5.20 muM than that of ABT at 74.7-460 muM. atipamezole 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30690186-9 2019 Atipamezole has more effective inhibition to CYP2C9 mediated diclofenac hydroxylation in human and animal liver microsomes with IC50 values of 1.50-5.20 muM than that of ABT at 74.7-460 muM. Diclofenac 61-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30690186-9 2019 Atipamezole has more effective inhibition to CYP2C9 mediated diclofenac hydroxylation in human and animal liver microsomes with IC50 values of 1.50-5.20 muM than that of ABT at 74.7-460 muM. 1-aminobenzotriazole 170-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30617764-11 2019 CONCLUSION: The presence of mutations in VKORC1 or CYP2C9 is associated with increased risk of bleeding in patients with BCS on warfarin. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 30088221-0 2019 Effect of Fluconazole Coadministration and CYP2C9 Genetic Polymorphism on Siponimod Pharmacokinetics in Healthy Subjects. siponimod 74-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 30617764-0 2019 Mutations in CYP2C9 and/or VKORC1 haplotype are associated with higher bleeding complications in patients with Budd-Chiari syndrome on warfarin. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 30088221-1 2019 OBJECTIVES: The aim of this study was to assess the pharmacokinetics (PK) and safety/tolerability of siponimod in healthy subjects when coadministered with (1) the moderate cytochrome P450 (CYP) 2C9 and CYP3A inhibitor fluconazole (Study A), and (2) with three different CYP2C9 genotype variants (Study B). siponimod 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-198 30088221-1 2019 OBJECTIVES: The aim of this study was to assess the pharmacokinetics (PK) and safety/tolerability of siponimod in healthy subjects when coadministered with (1) the moderate cytochrome P450 (CYP) 2C9 and CYP3A inhibitor fluconazole (Study A), and (2) with three different CYP2C9 genotype variants (Study B). siponimod 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 30088221-6 2019 In Study B, the AUCs of siponimod were approximately two to fourfold greater in subjects with the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes, with a minor increase in Cmax versus the CYP2C9*1/*1 genotype. siponimod 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 30088221-6 2019 In Study B, the AUCs of siponimod were approximately two to fourfold greater in subjects with the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes, with a minor increase in Cmax versus the CYP2C9*1/*1 genotype. siponimod 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 30088221-6 2019 In Study B, the AUCs of siponimod were approximately two to fourfold greater in subjects with the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes, with a minor increase in Cmax versus the CYP2C9*1/*1 genotype. siponimod 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 30088221-9 2019 CONCLUSIONS: Changes in siponimod PK, when coadministered with fluconazole at steady-state and in subjects with different CYP2C9 genotypes, indicate that the reduced CYP2C9 enzymatic activity does not affect the absorption phase of siponimod but prolongs the elimination phase. siponimod 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 30088221-9 2019 CONCLUSIONS: Changes in siponimod PK, when coadministered with fluconazole at steady-state and in subjects with different CYP2C9 genotypes, indicate that the reduced CYP2C9 enzymatic activity does not affect the absorption phase of siponimod but prolongs the elimination phase. siponimod 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 30088221-9 2019 CONCLUSIONS: Changes in siponimod PK, when coadministered with fluconazole at steady-state and in subjects with different CYP2C9 genotypes, indicate that the reduced CYP2C9 enzymatic activity does not affect the absorption phase of siponimod but prolongs the elimination phase. Fluconazole 63-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 30088221-10 2019 These results confirm the relevance of CYP2C9 activity on siponimod metabolism in humans. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 30617764-2 2019 Vitamin K epoxide reductase complex 1 (VKORC1) and CYP2C9 are involved in the metabolism of warfarin. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 30823561-5 2019 The metabolism of 25B-NBF was catalyzed by CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2J2, CYP3A4, and UGT2B7 enzymes. SSW3KY7SWW 18-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30883300-4 2019 The difference in patients" pharmacodynamic responses to warfarin has been attributed to genetic variation in warfarin metabolism and molecular targets (e.g., CYP2C9 and VKORC1) and host-environment interactions. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 30883300-4 2019 The difference in patients" pharmacodynamic responses to warfarin has been attributed to genetic variation in warfarin metabolism and molecular targets (e.g., CYP2C9 and VKORC1) and host-environment interactions. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 30883300-12 2019 CYP2C9*5, *8, *9, and *11, and VKORC1 Asp36Tyr genetic variants explained warfarin variability in Africans better, compared to CYP2C9*2 and *3. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30443705-0 2019 Non randomized study on the potential of nitisinone to inhibit cytochrome P450 2C9, 2D6, 2E1 and the organic anion transporters OAT1 and OAT3 in healthy volunteers. nitisinone 41-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-82 30443705-1 2019 PURPOSE: Nitisinone inhibits the cytochrome P450 (CYP) subfamilies CYP2C9, CYP2D6, and CYP2E1 and the organic anion transporter (OAT) isoforms OAT1 and OAT3 in vitro. nitisinone 9-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30443705-11 2019 Nitisinone was a moderate inhibitor of CYP2C9, and treatment may therefore result in increased plasma concentrations of comedications metabolized primarily via this enzyme. nitisinone 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 30472588-2 2019 In this study, we used a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and N-acetyltransferase-2 (NAT2) in plasma. Dapsone 61-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 30899365-3 2019 In TERT-HNECs treated with Y27632 for 5 days, upregulation of p63, gap junction molecules Cx26, Cx30, Cx43, cytochrome P450 enzymes CYP2C9, CYP2C18, CYP39A1, CYP4B1, CYP2G1P, CYP4Z1, and KLF families KLF10 and KLF11 were observed compared to the control. Y 27632 27-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 30472588-2 2019 In this study, we used a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and N-acetyltransferase-2 (NAT2) in plasma. Bupropion 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Bupropion 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Losartan 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 30472588-14 2019 CONCLUSIONS: This study advocates the use of a cocktail of bupropion, losartan and dapsone for in vivo phenotyping of CYP2B6, CYP2C9 and NAT2, which is important in determining therapeutic dose levels of anti-HIV and anti-TB drugs in HIV/AIDS-TB coinfections. Dapsone 83-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 30270535-0 2019 HLA Alleles and CYP2C9*3 as Predictors of Phenytoin Hypersensitivity in East Asians. Phenytoin 42-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 30466000-6 2019 Moreover, RA weakly inhibited CYP2C9 and 2E1 activities with IC50 values of 39.6 and 61.0 muM, respectively, while moderately inhibiting UGT1A1, 1A6, and 2B7 with IC50 values of 9.24, 19.1, and 23.4 muM, respectively. rosmarinic acid 10-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 30447161-6 2019 CONCLUSIONS AND IMPLICATIONS: The combined results show that the 5"-hydroxylation of flucloxacillin is primarily catalysed by CYP3A4, CYP3A7 and CYP2C9. Floxacillin 85-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 30270535-2 2019 In addition to cytochrome P450 (CYP)2C9*3, we found that HLA-B*13:01, HLA-B*15:02, and HLA-B*51:01 were significantly associated with phenytoin hypersensitivity with distinct phenotypic specificities. Phenytoin 134-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-39 30270535-3 2019 Strikingly, we showed an increase in predictive sensitivity of concurrently testing CYP2C9*3/HLA-B*13:01/HLA-B*15:02/HLA-B*51:01 from 30.5-71.9% for selecting the individuals with the risk of developing phenytoin-SCAR in Taiwanese cohorts, accompanied by a specificity of 77.7% (combined sensitivity, 64.7%; specificity, 71.9% for three Asian populations). Phenytoin 203-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 30270535-5 2019 In conclusion, combining the assessment of risk alleles of HLA and CYP2C9 potentiated the usefulness of predictive genetic tests to prevent phenytoin hypersensitivity in Asians. Phenytoin 140-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30121860-0 2019 Height, VKORC1 1173, and CYP2C9 Genotypes Determine Warfarin Dose for Pediatric Patients with Kawasaki Disease in Southwest China. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 30159654-0 2019 Effect of CYP2C9 Polymorphisms on the Pharmacokinetics of Indomethacin During Pregnancy. Indomethacin 58-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30159654-1 2019 BACKGROUND AND OBJECTIVE: Cytochrome P450 (CYP) 2C9 catalyzes the biotransformation of indomethacin to its inactive metabolite O-desmethylindomethacin (DMI). Indomethacin 87-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-51 30159654-1 2019 BACKGROUND AND OBJECTIVE: Cytochrome P450 (CYP) 2C9 catalyzes the biotransformation of indomethacin to its inactive metabolite O-desmethylindomethacin (DMI). 5-hydroxyindomethacin 127-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-51 30159654-1 2019 BACKGROUND AND OBJECTIVE: Cytochrome P450 (CYP) 2C9 catalyzes the biotransformation of indomethacin to its inactive metabolite O-desmethylindomethacin (DMI). 5-hydroxyindomethacin 152-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-51 30159654-2 2019 The aim of this work was to determine the effect of CYP2C9 polymorphisms on indomethacin metabolism in pregnant women. Indomethacin 76-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 30159654-10 2019 CONCLUSION: Although our results are limited by sample size and are not statistically significant, these data suggest that certain genetic polymorphisms of CYP2C9 may lead to an increased metabolic ratio and an increase in the clearance of indomethacin. Indomethacin 240-252 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 30159654-11 2019 More data are needed to assess the impact of CYP2C9 genotype on the effectiveness of indomethacin as a tocolytic agent. Indomethacin 85-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30678826-9 2019 Interestingly, several metabolites including the 4-hydroxyl form of CPA (4-OH-CPA) and phosphamide mustard were detected in the CYP2B6, CYP2C19, and CYP3A4 expression systems, but not in the CYP2C9 and CYP2D6 system. 4-hydroxyl 49-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 30678826-9 2019 Interestingly, several metabolites including the 4-hydroxyl form of CPA (4-OH-CPA) and phosphamide mustard were detected in the CYP2B6, CYP2C19, and CYP3A4 expression systems, but not in the CYP2C9 and CYP2D6 system. Dimethoate 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 191-197 30678826-13 2019 Additionally, CPA metabolites like 4-OH-CPA and phosphamide mustard produced by human CYP2B6, CYP2C9, CYP2C19, and CYP3A4 are suggested to be major determinants of micronucleus induction by CPA. Dimethoate 48-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 29375004-5 2019 Contributions of CYP3A4 and CYP2C9 to metabolism of deoxypodophyllotoxin and nateglinide were also predicted. deoxypodophyllotoxin 52-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 29375004-8 2019 Predicted contributions of CYP3A4 and CYP2C9 to nateglinide metabolism were 8.18-37.84% and 36.08-94.04%, separately. Nateglinide 48-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 29382249-6 2019 Methoxsalen is a direct inhibitor and inhibited the activities (>90%) of all enzymes at a concentration of 300 microM except for CYP2C9. Methoxsalen 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 29382249-9 2019 ABT is a potent TDI of several P450 except for CYP2C19 (47%) and CYP2C9 (27%). 2,2'-azino-di-(3-ethylbenzothiazoline)-6-sulfonic acid 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 30326170-1 2019 Delta-9-tetrahydrocannabinol (THC), the main psychoactive cannabinoid in cannabis, may inhibit the cytochrome P450 enzyme CYP2C9. Dronabinol 0-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 30326170-1 2019 Delta-9-tetrahydrocannabinol (THC), the main psychoactive cannabinoid in cannabis, may inhibit the cytochrome P450 enzyme CYP2C9. Dronabinol 30-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 30326170-1 2019 Delta-9-tetrahydrocannabinol (THC), the main psychoactive cannabinoid in cannabis, may inhibit the cytochrome P450 enzyme CYP2C9. Cannabinoids 58-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 30326170-6 2019 The in vitro study indicated that THC inhibits the CYP2C9-mediated metabolism of warfarin. Dronabinol 34-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 30326170-6 2019 The in vitro study indicated that THC inhibits the CYP2C9-mediated metabolism of warfarin. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 30336288-11 2019 Identification of orthologous human CYP2C9 and CYP3A4 enzyme families in vultures will be an important further step in explaining the differences in the metabolic pathway(s) of meloxicam and diclofenac for the species. Meloxicam 177-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 30336288-11 2019 Identification of orthologous human CYP2C9 and CYP3A4 enzyme families in vultures will be an important further step in explaining the differences in the metabolic pathway(s) of meloxicam and diclofenac for the species. Diclofenac 191-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 29119932-0 2019 Relevance of CYP2C9 Function in Valproate Therapy. Valproic Acid 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 29119932-4 2019 METHODS: CYP2C9 mediated oxidation of valproate, which is the minor metabolic pathway in adults, appears to become the principal route in children. Valproic Acid 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 29119932-5 2019 Genetic and non-genetic variations in CYP2C9 activity can result in significant inter- and intra-individual differences in valproate pharmacokinetics and valproate induced adverse reactions. Valproic Acid 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 29119932-5 2019 Genetic and non-genetic variations in CYP2C9 activity can result in significant inter- and intra-individual differences in valproate pharmacokinetics and valproate induced adverse reactions. Valproic Acid 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 94-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 94-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 94-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 94-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 229-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 229-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 229-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-6 2019 RESULTS: The loss-of-function alleles, CYP2C9*2 or CYP2C9*3, display significant reduction in valproate metabolism in children; furthermore, low CYP2C9 expression in patients with CYP2C9*1/*1 genotype also leads to a decrease in valproate metabolizing capacity. Valproic Acid 229-238 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29119932-8 2019 CONCLUSION: Novel strategy for valproate therapy adjusted to CYP2C9-status (CYP2C9 genotype and CYP2C9 expression) is strongly recommended in childhood. Valproic Acid 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29119932-8 2019 CONCLUSION: Novel strategy for valproate therapy adjusted to CYP2C9-status (CYP2C9 genotype and CYP2C9 expression) is strongly recommended in childhood. Valproic Acid 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 29119932-8 2019 CONCLUSION: Novel strategy for valproate therapy adjusted to CYP2C9-status (CYP2C9 genotype and CYP2C9 expression) is strongly recommended in childhood. Valproic Acid 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 29119932-9 2019 The early knowledge of pediatric patients" CYP2C9-status facilitates the optimization of valproate dosing which contributes to the avoidance of misdosing induced adverse reactions, such as abnormal blood levels of ammonia and alkaline phosphatase, and improves the safety of children"s anticonvulsant therapy. Valproic Acid 89-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 30566377-2 2019 The rs12777823G>A single-nucleotide polymorphism (SNP) in the CYP2C gene cluster has been shown to influence optimal warfarin doses in African Americans. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-70 30566377-11 2019 Given ongoing worldwide efforts to identify clinically relevant human genetic variation impacting on optimal warfarin dose selection, the African ancestry-specific genetic variant in the CYP2C cluster and others warrant further research and consideration in development of future warfarin dosing algorithms for precision medicine guidelines. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-192 30566377-11 2019 Given ongoing worldwide efforts to identify clinically relevant human genetic variation impacting on optimal warfarin dose selection, the African ancestry-specific genetic variant in the CYP2C cluster and others warrant further research and consideration in development of future warfarin dosing algorithms for precision medicine guidelines. Warfarin 280-288 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 187-192 30584236-2 2018 This study purposed to investigate the effect of OATP1B3 and CYP2C9 genetic polymorphisms on the transport and metabolism of glibenclamide and glipizide in human. Glyburide 125-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 30518301-0 2019 CYP2C9*61, a rare missense variant identified in a Puerto Rican patient with low warfarin dose requirements. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30518301-3 2019 This case report describes a rare missense variant at exon 9 of CYP2C9 (rs202201137; c.1370A>G transition; p.Asn457Ser) found in a Puerto Rican patient with low warfarin dose requirements (3 mg/day). Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 30518301-4 2019 The haplotype characterized by two amino acid changes, Asn457Ser and Arg144Cys (rs1799853; c.430C>T), has been designated CYP2C9*61 by the Pharmacogene Variation Consortium. asn457ser 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 30584236-0 2018 OATP1B3 (699G>A) and CYP2C9*2, *3 significantly influenced the transport and metabolism of glibenclamide and glipizide. Glyburide 94-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 29297772-6 2019 CC-223 was metabolized by CYP2C9 and CYP3A, while metabolism of M1 was mediated by CYP2C8 and CYP3A. CC-223 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 30584236-2 2018 This study purposed to investigate the effect of OATP1B3 and CYP2C9 genetic polymorphisms on the transport and metabolism of glibenclamide and glipizide in human. Glipizide 143-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 30584236-0 2018 OATP1B3 (699G>A) and CYP2C9*2, *3 significantly influenced the transport and metabolism of glibenclamide and glipizide. Glipizide 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 30584236-3 2018 An LC-MS method was used to determine the uptake of glibenclamide and glipizide in OATP1B3, OATP1B3 (344T > G) and OATP1B3 (699G > A)-HEK293T cells and their metabolism in CYP2C9*1, *2 and *3 recombinase system. Glyburide 52-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 30584236-6 2018 Glibenclamide can be metabolized in CYP2C9*1, *2 and *3 recombinase system with the Vmax values of 1.58 +- 0.71, 0.69 +- 0.25, and 0.41 +- 0.13 nmol/min/mg protein, while glipizide was metabolized with Vmax of 8.82 +- 2.78, 5.99 +- 1.95, and 2.87 +- 1.03 nmol/min/mg protein, respectively. Glyburide 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 30584236-6 2018 Glibenclamide can be metabolized in CYP2C9*1, *2 and *3 recombinase system with the Vmax values of 1.58 +- 0.71, 0.69 +- 0.25, and 0.41 +- 0.13 nmol/min/mg protein, while glipizide was metabolized with Vmax of 8.82 +- 2.78, 5.99 +- 1.95, and 2.87 +- 1.03 nmol/min/mg protein, respectively. Glipizide 171-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 30584236-7 2018 The internal clearance of glibenclamide and glipizide in CYP2C9*2 and *3 was markedly reduced compared to that in CYP2C9*1. Glyburide 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 30584236-7 2018 The internal clearance of glibenclamide and glipizide in CYP2C9*2 and *3 was markedly reduced compared to that in CYP2C9*1. Glyburide 26-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 30584236-7 2018 The internal clearance of glibenclamide and glipizide in CYP2C9*2 and *3 was markedly reduced compared to that in CYP2C9*1. Glipizide 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 30584236-8 2018 These results collectively demonstrate that OATP1B3 (699G > A) and CYP2C9*2 and *3 have a significant influence on the transport and metabolism of glibenclamide and glipizide. Glyburide 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 30584236-8 2018 These results collectively demonstrate that OATP1B3 (699G > A) and CYP2C9*2 and *3 have a significant influence on the transport and metabolism of glibenclamide and glipizide. Glipizide 168-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 30412386-8 2018 Kinetic analyses using recombinant proteins revealed that CYP2C9 variants (A82V and H344R) showed substantially lower Ks values than the wild type, although CYP1A1 variant (V382I) showed kinetic parameters similar to the wild type. Potassium 119-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 30105453-0 2018 Siponimod pharmacokinetics, safety, and tolerability in combination with rifampin, a CYP2C9/3A4 inducer, in healthy subjects. siponimod 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 29744741-6 2018 Furthermore, 1 microM naltrexone hydrochloride inhibited CYP3A4 enzyme activity, the most by 37.9% followed by CYP2C9 (36.5%) and CYP2D6 (31.8%). Naltrexone 22-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 29744741-7 2018 The CYP2C9 and CYP2D6 metabolic activities were greatly affected by naltrexone hydrochloride, which even at the lowest concentration of naltrexone hydrochloride (0.01 microM) significantly decreased the metabolic activity by 34.9 and 16.0%, respectively. Naltrexone 68-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 29744741-7 2018 The CYP2C9 and CYP2D6 metabolic activities were greatly affected by naltrexone hydrochloride, which even at the lowest concentration of naltrexone hydrochloride (0.01 microM) significantly decreased the metabolic activity by 34.9 and 16.0%, respectively. Naltrexone 136-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 29744741-9 2018 CONCLUSION: These outcomes advocate that there is a great possibility of drug interactions resulting from the concurrent administration of naltrexone hydrochloride with actives that are metabolized by these CYP enzymes, particularly CYP2C9 and CYP2D6. Naltrexone 139-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 233-239 30105453-1 2018 PURPOSE: To assess the potential pharmacokinetic (PK) interactions between siponimod and rifampin, a strong CYP3A4/moderate CYP2C9 inducer, in healthy subjects. siponimod 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 30105453-1 2018 PURPOSE: To assess the potential pharmacokinetic (PK) interactions between siponimod and rifampin, a strong CYP3A4/moderate CYP2C9 inducer, in healthy subjects. Rifampin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 30105453-14 2018 CONCLUSIONS: The study findings suggest that in the presence of rifampin, a strong CYP3A4/moderate CYP2C9 inducer, siponimod showed significant decrease in Cmax,ss (45%) and AUCtau,ss (57%) in healthy subjects. Rifampin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 29569712-1 2018 Rifampin demonstrated dose-dependent relative induction between cytochrome P (CYP)3A and P-glycoprotein (P-gp), organic anion transporting polypeptides (OATPs), or CYP2C9; P-gp, OATP, and CYP2C9 induction was one drug-drug interaction (DDI) category lower than that observed for CYP3A across a wide range of pregnane X receptor (PXR) agonism. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 29633238-0 2018 Drinking Ethanol Has Few Acute Effects on CYP2C9, CYP2C19, NAT2, and P-Glycoprotein Activities but Somewhat Inhibits CYP1A2, CYP2D6, and Intestinal CYP3A: So What? Ethanol 9-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 29905956-0 2018 Population Pharmacokinetic/Pharmacodynamic Analyses of Avatrombopag in Patients With Chronic Liver Disease and Optimal Dose Adjustment Guide With Concomitantly Administered CYP3A and CYP2C9 Inhibitors. avatrombopag 55-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 183-189 30595245-0 2018 Association between genetic polymorphisms of CYP2C9 and VKORC1 and safety and efficacy of warfarin: Results of a 5 years audit. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30595245-1 2018 OBJECTIVE: Genetic polymorphisms of CYP2C9 and VKORC1 play major role in pharmacokinetics and pharmacodynamics of warfarin, respectively. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 30179260-7 2018 Additionally, nilotinib is a competitive inhibitor of CYP3A4/5, CYP2C8, CYP2C9, CYP2D6, and uridine diphosphate glucuronosyltransferase 1A1. nilotinib 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 29802808-0 2018 CYP2C9*3 gene variant contributes independently to glycaemic control in patients with type 2 diabetes treated with glibenclamide. Glyburide 115-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30142539-0 2018 Interactions of casticin, ipriflavone, and resveratrol with serum albumin and their inhibitory effects on CYP2C9 and CYP3A4 enzymes. casticin 16-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 29802808-2 2018 In this cross-sectional study, the aim was to explore the association of genetic variants in CYP2C9, ABCC8, KCNJ11 and TCF7L2 with good glycaemic control in Mexican type 2 diabetes (T2D) treated with glibenclamide. Glyburide 200-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 29802808-11 2018 WHAT IS NEW AND CONCLUSION: The findings suggest that CYP2C9*3 genetic variant independently contributes to good glycaemic control of patients with type 2 diabetes treated with glibenclamide. Glyburide 177-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Methadone 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 30205091-2 2018 Methadone undergoes N-demethylation by multiple cytochrome P450 (CYP) enzymes including CYP3A4, CYP2B6, CYP2C19, CYP2D6, CYP2C9, and CYP2C8. Nitrogen 20-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 30363031-1 2018 OBJECTIVE: The aim of this study was to determine the impact of the SLCO1B1, apolipoprotein E (ApoE), and CYP2C9 genotypes on the lipid-lowering efficacy of fluvastatin. Fluvastatin 157-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 30486437-0 2018 Impact of CYP2C9 and VKORC1 Polymorphisms on Warfarin Sensitivity and Responsiveness in Jordanian Cardiovascular Patients during the Initiation Therapy. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30486437-4 2018 This study confirmed a genetic association of the CYP2C9*3 and VKORC1 rs10871454, rs8050894, rs9934438, and rs17708472 SNPs with warfarin sensitivity. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 30486437-5 2018 This study also found an association between CYP2C9 and VKORC1 genetic haplotype blocks and warfarin sensitivity. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 30486437-6 2018 The initial warfarin dose was significantly related to the CYP2C9*3 polymorphism and the four VKORC1 SNPs (p < 0.001). Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 30486437-7 2018 There were significant associations between rs4086116 SNP and TAT haplotype within CYP2C9 gene and rs17708472 SNP and CCGG haplotype within VKORC1 gene and warfarin responsiveness. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 30363031-0 2018 The association between the SLCO1B1, apolipoprotein E, and CYP2C9 genes and lipid response to fluvastatin: a meta-analysis. Fluvastatin 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 30452466-9 2018 Among loci which have extraordinary MAFs in Roma population two have strong proof of clinical importance: rs1799853 (CYP2C9) for warfarin dosage, and rs12248560 (CYP2C19) for clopidogrel dosage, efficacy and toxicity. Warfarin 129-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 30452466-9 2018 Among loci which have extraordinary MAFs in Roma population two have strong proof of clinical importance: rs1799853 (CYP2C9) for warfarin dosage, and rs12248560 (CYP2C19) for clopidogrel dosage, efficacy and toxicity. Clopidogrel 175-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 30142539-8 2018 Our results suggest that high intake of CAS and RES may interfere with the albumin-binding of Site II ligands as well as the metabolism of drugs by CYP2C9 and/or CYP3A4 enzymes, while large doses of IPR may affect the CYP3A4-catalyzed biotransformation of some drugs. casticin 40-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 30142539-8 2018 Our results suggest that high intake of CAS and RES may interfere with the albumin-binding of Site II ligands as well as the metabolism of drugs by CYP2C9 and/or CYP3A4 enzymes, while large doses of IPR may affect the CYP3A4-catalyzed biotransformation of some drugs. Resveratrol 48-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 30142539-6 2018 Furthermore, the effects of CAS, IPR, and RES on CYP2C9 and CYP3A4 enzymes were examined, employing diclofenac and testosterone as substrates, respectively. Resveratrol 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 30142539-7 2018 Our main observations are the following: (1) Polyphenols formed stable complexes with albumin (K = 104-105 L/mol); (2) CAS and RES slightly displaced naproxen from human albumin, while albumin-binding of warfarin was not affected; (3) CAS and RES significantly inhibited CYP2C9, with CAS being as potent as the positive control warfarin; (4) each polyphenol significantly inhibited CYP3A4, with RES being stronger and CAS slightly weaker than the known inhibitor naringenin. Polyphenols 45-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 30362120-5 2018 Inhibition studies with chemical inhibitors and antibodies suggested that arachidonic acid, the substrate of CYP2J2, and CYP2J2-specific antibody effectively inhibited the formation of SYL-927-M in HLMs whereas no significant inhibition was observed with the inhibitors for CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4, demonstrating that CYP2J2 was primarily responsible for the formation of SYL-927-M. Arachidonic Acid 74-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 290-296 30142539-7 2018 Our main observations are the following: (1) Polyphenols formed stable complexes with albumin (K = 104-105 L/mol); (2) CAS and RES slightly displaced naproxen from human albumin, while albumin-binding of warfarin was not affected; (3) CAS and RES significantly inhibited CYP2C9, with CAS being as potent as the positive control warfarin; (4) each polyphenol significantly inhibited CYP3A4, with RES being stronger and CAS slightly weaker than the known inhibitor naringenin. casticin 119-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 30138636-6 2018 On CYP2C9, apatinb exhibited the noncompetitive inhibition (Ki = 0.71 muM) while it inhibited CYP2C11 uncompetitively (Ki = 3.30 muM). apatinb 11-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 30345882-4 2018 Observational studies have demonstrated the significant impact of single nucleotide polymorphisms in CYP2C9 and VKORC1 on warfarin dose in patients of European ancestry and African-Americans. Warfarin 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 30321703-5 2018 RESULTS: S-warfarin clearance was significantly lower in patients with either CYP2C9 single (*2 or *3) or double (*2*2 or *2*3) variant alleles compared to those with wild-type genotype (P < 0.001). Warfarin 11-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 30321703-6 2018 Regression analysis revealed that patient age (P = 0.037) and CYP2C9 *2*2 & *2*3 genotype (P = 0.005), but not VKORC1 genotype, significantly affected the time taken for the resumption of normal coagulation (INR value declining to <=1.5). Adenosine Monophosphate 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 30099291-0 2018 Effects of dibutyl phthalate and di(2-ethylhexyl) phthalate with their metabolites on CYP2C9*1 and CYP2C19*1 activities in vitro. Dibutyl Phthalate 11-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 30321703-7 2018 CONCLUSIONS: The inter-individual variability in the time needed for normal coagulation to resume following warfarin withdrawal is influenced, in the main, by variance in S-warfarin clearance, which in turn is affected by CYP2C9 polymorphism and age. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 30321703-7 2018 CONCLUSIONS: The inter-individual variability in the time needed for normal coagulation to resume following warfarin withdrawal is influenced, in the main, by variance in S-warfarin clearance, which in turn is affected by CYP2C9 polymorphism and age. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 30321703-8 2018 Cost-effectiveness of pharmacogenetics-based algorithms incorporating CYP2C9 genotype and patient age could be increased if used not only to guide dosing decisions but also estimation of the correct length of time needed for individual patients to stop taking warfarin prior to surgery. Warfarin 260-268 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 30099291-0 2018 Effects of dibutyl phthalate and di(2-ethylhexyl) phthalate with their metabolites on CYP2C9*1 and CYP2C19*1 activities in vitro. Diethylhexyl Phthalate 33-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 30099291-1 2018 The aim of this article is to assess the effect of phthalates on the activities of CYP2C9 and CYP2C19 in vitro. phthalic acid 51-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 30099291-4 2018 The results showed dibutyl phthalate (DBP) significantly inhibited CYP2C9*1 with an activity inhibition rate of 67.3% and half-maximal inhibitory concentration (IC50) of 29.63 mumol L-1, but its metabolite monobutyl phthalate (MBP) had no significant effect. Dibutyl Phthalate 19-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30099291-4 2018 The results showed dibutyl phthalate (DBP) significantly inhibited CYP2C9*1 with an activity inhibition rate of 67.3% and half-maximal inhibitory concentration (IC50) of 29.63 mumol L-1, but its metabolite monobutyl phthalate (MBP) had no significant effect. Dibutyl Phthalate 38-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30099291-8 2018 Further investigation showed MEHP was a competitive inhibitor of CYP2C9*1 (Ki = 7.063 mumol L-1), and DBP was a competitive inhibitor of CYP2C19*1 (Ki = 7.013 mumol L-1) against their substrates, both phthalates were non-competitive inhibitors of the cofactor NADPH. mono-(2-ethylhexyl)phthalate 29-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 30099291-9 2018 Our results suggested that DBP, DEHP, and their metabolites exhibited significant inhibitory effects toward either CYP2C9*1 or CYP2C19*1. Diethylhexyl Phthalate 32-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 30356105-6 2018 Similarly, South Asians had a significantly higher frequency of CYP2C9*3 (12% SDS, 13% GIH, 11% SAS) vs. 7% in Europeans (pgenotype = <1.0E-05) and "T" allele of CYP4F2 (36%) SDS, (43%) GIH, 40% (SAS) vs. (29%) in Europeans (pgenotype = <1.0E-05); both associated with a higher risk of bleeding with warfarin. sds 78-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 30339708-3 2018 Remarkable efforts have been made to develop the machine learning based warfarin dosing algorithms incorporating clinical factors and genetic variants such as polymorphisms in CYP2C9 and VKORC1. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 30410385-3 2018 CYP2C9 and VKORC1 gene polymorphisms affect warfarin metabolism and dosage. Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30410385-8 2018 Results: Median weekly prescribed warfarin dosage was significantly lower in patients having the VKORC1-1639 AA genotype and poor CYP2C9*2/*2,*2/*3 metabolizers than their wild-type counterparts. Warfarin 34-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 29994939-3 2018 The recent pharmacogenetics studies revealed an association of the organic cation transporter 1 (OCT1) and ATP-binding cassette C3 (ABCC3) polymorphisms with morphine-related adverse effects, an effect of polymorphisms in cytochrome P450 gene CYP2D6 on the analgesic efficacy of tramadol and no effect of CYP2C8 and CYP2C9 variants on efficacy of piroxicam. Morphine 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 316-322 30010032-7 2018 Conversely, the inhibitory ratios of 3,4-DCPB (16 mug/mL, 59 muM) on the activities of CYP2C9, CYP2C19, CYP1A2 and CYP2D6 were 97.6%, 59.0%, 53.5% and 36.5%, respectively. 3,4-dichlorophenyl-propenoyl-sec-butylamine 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 30010032-10 2018 3,4-DCPB inhibited the activities of CYP2C9, CYP2C19, CYP1A2 and CYP2D6, rather than that CYP2E1 and CYP3A4 enzymes, suggesting that the different effects of 3,4-DCPB on the CYP450 enzymes might influence metabolic drug-drug interaction in antiepileptics therapy. 3,4-dichlorophenyl-propenoyl-sec-butylamine 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 30010032-10 2018 3,4-DCPB inhibited the activities of CYP2C9, CYP2C19, CYP1A2 and CYP2D6, rather than that CYP2E1 and CYP3A4 enzymes, suggesting that the different effects of 3,4-DCPB on the CYP450 enzymes might influence metabolic drug-drug interaction in antiepileptics therapy. 3,4-dichlorophenyl-propenoyl-sec-butylamine 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 29958892-4 2018 RESULTS: In this case we present a patient with multifactorial vitamin K deficiency (due to nutritional defects and multiple genetic mutations in VKOR and CYP2C9) that was exacerbated by antibiotic and warfarin therapy during her hospital admission. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 30319126-7 2018 Moreover, enzalutamide may require dose adjustment for other drugs since it is a potent inductor of CYP3A4 and a moderate inductor of CYP2C9 y el CYP2C19. enzalutamide 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 30219715-4 2018 In individual HLMs with variable CYP2D6 activities, a significant correlation was observed between celecoxib hydroxylation and CYP2D6-selective dextromethorphan O-demethylation when CYP2C9 and CYP3A4 activities were suppressed (r = 0.97, P < 0.0001). Celecoxib 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 30219715-4 2018 In individual HLMs with variable CYP2D6 activities, a significant correlation was observed between celecoxib hydroxylation and CYP2D6-selective dextromethorphan O-demethylation when CYP2C9 and CYP3A4 activities were suppressed (r = 0.97, P < 0.0001). Dextromethorphan 144-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 30219715-8 2018 Using HLMs from individual donors, the relative contribution of CYP2D6 to celecoxib metabolism was found to be highly variable and dependent on CYP2C9 genotypes, ranging from no contribution in extensive metabolizers with CYP2C9*1*1 genotype to approximately 30% in slow metabolizers with allelic variants CYP2C9*1*3 and CYP2C9*3*3. Celecoxib 74-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 30219715-9 2018 These results demonstrate that celecoxib may become a potential victim of CYP2D6-associated drug-drug interactions, particularly in individuals with reduced CYP2C9 activity. Celecoxib 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 30219715-0 2018 Celecoxib is a substrate of CYP2D6: Impact on celecoxib metabolism in individuals with CYP2C9*3 variants. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 30219715-0 2018 Celecoxib is a substrate of CYP2D6: Impact on celecoxib metabolism in individuals with CYP2C9*3 variants. Celecoxib 46-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 30219715-3 2018 In human liver microsomes (HLMs), a concentration-dependent inhibition of celecoxib hydroxylation by quinidine was observed after CYP2C9 and CYP3A4 were inhibited. Celecoxib 74-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 30219715-3 2018 In human liver microsomes (HLMs), a concentration-dependent inhibition of celecoxib hydroxylation by quinidine was observed after CYP2C9 and CYP3A4 were inhibited. Quinidine 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 30283820-0 2018 DRESS after IV phenytoin associated with cytochrome P450 CYP2C9*3 homozygosity. Phenytoin 15-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 30098040-0 2018 Metabolism of steroids by cytochrome P450 2C9 variants. Steroids 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-45 30178440-0 2018 The influences of CYP2C9*1/*3 genotype on the pharmacokinetics of zolpidem. Zolpidem 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 29916837-7 2018 To explain the association between demographic, clinical and genotype data (VKORC1, CYP2C9 and CYP4F2) and the mean weekly dose of acenocoumarol, we performed a multiple linear regression model. Acenocoumarol 131-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 30012669-0 2018 The Cytochrome P450 Slow Metabolizers CYP2C9*2 and CYP2C9*3 Directly Regulate Tumorigenesis via Reduced Epoxyeicosatrienoic Acid Production. epoxyeicosatrienoic acid 104-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 30012669-0 2018 The Cytochrome P450 Slow Metabolizers CYP2C9*2 and CYP2C9*3 Directly Regulate Tumorigenesis via Reduced Epoxyeicosatrienoic Acid Production. epoxyeicosatrienoic acid 104-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 30012669-1 2018 Increased expression of cytochrome P450 CYP2C9, together with elevated levels of its products epoxyeicosatrienoic acids (EET), is associated with aggressiveness in cancer. epoxyeicosatrienoic acids 94-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 30012669-1 2018 Increased expression of cytochrome P450 CYP2C9, together with elevated levels of its products epoxyeicosatrienoic acids (EET), is associated with aggressiveness in cancer. EET 121-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 30012669-3 2018 Although genetic differences in CYP2C9-dependent oxidation of arachidonic acid (AA) have been reported, the roles of CYP2C9*2 and CYP2C9*3 in EET biosynthesis and their relevance to disease are unknown. Arachidonic Acid 62-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 30012669-5 2018 When injected into mice, NSCLC cells expressing CYP2C9*2 and CYP2C9*3 produced lower levels of EETs and developed fewer, smaller, and less vascularized tumors than cells expressing CYP2C9*1. eets 95-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 30012669-5 2018 When injected into mice, NSCLC cells expressing CYP2C9*2 and CYP2C9*3 produced lower levels of EETs and developed fewer, smaller, and less vascularized tumors than cells expressing CYP2C9*1. eets 95-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 30012669-5 2018 When injected into mice, NSCLC cells expressing CYP2C9*2 and CYP2C9*3 produced lower levels of EETs and developed fewer, smaller, and less vascularized tumors than cells expressing CYP2C9*1. eets 95-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 30012669-7 2018 Purified CYP2C9*2 and CYP2C9*3 exhibited attenuated catalytic efficiency in producing EETs, primarily due to impaired reduction of these two variants by NADPH-P450 reductase. eets 86-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 30012669-7 2018 Purified CYP2C9*2 and CYP2C9*3 exhibited attenuated catalytic efficiency in producing EETs, primarily due to impaired reduction of these two variants by NADPH-P450 reductase. eets 86-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 28992765-6 2018 The major isoforms of CYP involved in the metabolism of warfarin sodium are CYP1A2 (for the R-warfarin) and CYP2C9 (for the S-warfarin). Warfarin 56-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 28992765-6 2018 The major isoforms of CYP involved in the metabolism of warfarin sodium are CYP1A2 (for the R-warfarin) and CYP2C9 (for the S-warfarin). Warfarin 92-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 28992765-6 2018 The major isoforms of CYP involved in the metabolism of warfarin sodium are CYP1A2 (for the R-warfarin) and CYP2C9 (for the S-warfarin). Sulfur 124-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 28992765-6 2018 The major isoforms of CYP involved in the metabolism of warfarin sodium are CYP1A2 (for the R-warfarin) and CYP2C9 (for the S-warfarin). Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 28992765-7 2018 The probes for testing CYP1A2 are phenacetin and caffeine while for CYP2C9 tolbutamide. Tolbutamide 75-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 29756176-0 2018 Response: Similar effect of quercetin on CYP2E1 and CYP2C9 activities in humans? Quercetin 28-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 29770840-0 2018 Similar effect of quercetin on CYP2E1 and CYP2C9 activities in humans? Quercetin 18-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 30178440-1 2018 Zolpidem is predominantly metabolized by CYP3A4, and to a lesser extent by CYP2C9, CYP1A2, CYP2D6 and CYP2C19. Zolpidem 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 30178440-2 2018 The aim of this study was to identify the effects of CYP2C9*3 allele on the pharmacokinetics of zolpidem. Zolpidem 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 30178440-5 2018 In addition, since zolpidem is metabolized at a high rate by CYP3A4, the effect of CYP2C9*3 allele on the pharmacokinetics of zolpidem was also observed in the condition where CYP3A4 was sufficiently inhibited by the steady-state concentration of clarithromycin, a potent CYP3A4 inhibitor. Zolpidem 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30178440-5 2018 In addition, since zolpidem is metabolized at a high rate by CYP3A4, the effect of CYP2C9*3 allele on the pharmacokinetics of zolpidem was also observed in the condition where CYP3A4 was sufficiently inhibited by the steady-state concentration of clarithromycin, a potent CYP3A4 inhibitor. Zolpidem 126-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 30178440-5 2018 In addition, since zolpidem is metabolized at a high rate by CYP3A4, the effect of CYP2C9*3 allele on the pharmacokinetics of zolpidem was also observed in the condition where CYP3A4 was sufficiently inhibited by the steady-state concentration of clarithromycin, a potent CYP3A4 inhibitor. Clarithromycin 247-261 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 29859403-3 2018 Azoles" affinities toward human cytochrome P450 isoforms involved in xenobiotic metabolism (CYP3A4, CYP2C9 and CYP2D6) as well as fungal CYP51 were estimated with a ligand-protein docking model based on the ChemScore scoring function. Azoles 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 30098040-5 2018 CYP2C9.2, CYP2C9.3, CYP2C9.4, CYP2C9.16, CYP2C9.28, CYP2C9.48 and CYP2C9.52 had higher testosterone 6beta-hydroxylation than CYP2C9.1. testosterone 6beta 87-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-6 2018 CYP2C9.4 showed higher progesterone 6beta-hydroxylation activity than CYP2C9.1. Progesterone 23-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30098040-7 2018 CYP2C9.28 and CYP2C9.48 showed higher progesterone 11alpha-hydroxylation activity than CYP2C9.1. Progesterone 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30098040-7 2018 CYP2C9.28 and CYP2C9.48 showed higher progesterone 11alpha-hydroxylation activity than CYP2C9.1. Progesterone 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 30098040-7 2018 CYP2C9.28 and CYP2C9.48 showed higher progesterone 11alpha-hydroxylation activity than CYP2C9.1. Progesterone 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 30098040-9 2018 CYP2C9.2, CYP2C9.3, CYP2C9.16 and CYP2C9.30 had higher estrone 16alpha-hydroxylation activity than CYP2C9.1. Estrone 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30098040-9 2018 CYP2C9.2, CYP2C9.3, CYP2C9.16 and CYP2C9.30 had higher estrone 16alpha-hydroxylation activity than CYP2C9.1. Estrone 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30117082-1 2018 Zolpidem is indicated for the short-term treatment of insomnia and it is predominantly metabolized by CYP3A4, and to a lesser extent by CYP2C19, CYP1A2, and CYP2C9. Zolpidem 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 30098040-9 2018 CYP2C9.2, CYP2C9.3, CYP2C9.16 and CYP2C9.30 had higher estrone 16alpha-hydroxylation activity than CYP2C9.1. Estrone 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-9 2018 CYP2C9.2, CYP2C9.3, CYP2C9.16 and CYP2C9.30 had higher estrone 16alpha-hydroxylation activity than CYP2C9.1. Estrone 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-9 2018 CYP2C9.2, CYP2C9.3, CYP2C9.16 and CYP2C9.30 had higher estrone 16alpha-hydroxylation activity than CYP2C9.1. Estrone 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30098040-10 2018 CYP2C9.3 had higher estrone 11alpha-hydroxylation activity than CYP2C9.1. Estrone 20-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29741099-8 2018 There are suggestions that genotyping the CYP2C9 and VKORC1 genes is useful for determining a more appropriate initial dose and thereby increasing the effectiveness and safety of warfarin. Warfarin 179-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 29578591-5 2018 Danshensu and rosmarinic acid inhibited CYP2E1 and CYP2C9 with IC50 values of 36.63 and 75.76 mum, and 34.42 and 76.89 mum, respectively. 3,4-dihydroxyphenyllactic acid 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29578591-5 2018 Danshensu and rosmarinic acid inhibited CYP2E1 and CYP2C9 with IC50 values of 36.63 and 75.76 mum, and 34.42 and 76.89 mum, respectively. rosmarinic acid 14-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29578591-6 2018 Salvianolic acid A and B inhibited CYP2D6, CYP2E1 and CYP2C9 with IC50 values of 33.79, 21.64 and 31.94 mum, and 45.47, 13.52 and 24.15 mum, respectively. salvianolic acid A 0-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 29578591-6 2018 Salvianolic acid A and B inhibited CYP2D6, CYP2E1 and CYP2C9 with IC50 values of 33.79, 21.64 and 31.94 mum, and 45.47, 13.52 and 24.15 mum, respectively. Boron 23-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 29885857-3 2018 Gentisic acid (GA) is a minor catabolite of aspirin; yet humans carrying CYP2C9-variants incapable to catabolize aspirin to GA do not benefit from aspirin in prevention against colonic adenoma. 2,5-dihydroxybenzoic acid 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 29885857-3 2018 Gentisic acid (GA) is a minor catabolite of aspirin; yet humans carrying CYP2C9-variants incapable to catabolize aspirin to GA do not benefit from aspirin in prevention against colonic adenoma. Aspirin 113-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 29885857-3 2018 Gentisic acid (GA) is a minor catabolite of aspirin; yet humans carrying CYP2C9-variants incapable to catabolize aspirin to GA do not benefit from aspirin in prevention against colonic adenoma. 2,5-dihydroxybenzoic acid 124-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 29885857-3 2018 Gentisic acid (GA) is a minor catabolite of aspirin; yet humans carrying CYP2C9-variants incapable to catabolize aspirin to GA do not benefit from aspirin in prevention against colonic adenoma. Aspirin 113-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 29578591-4 2018 The results showed that DHI inhibited CYP2C19, CYP2D6, CYP3A4, CYP2E1 and CYP2C9 with IC50 values of 1.26, 1.42, 1.63, 1.10 and 1.67% (v/v), respectively. dehydrosoyasaponin I 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 30017171-2 2018 Co-administration with other medications can affect the cytochrome P450 (CYP) 2C9 enzyme function; thus, imrecoxib metabolism can be affected. Imrecoxib 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-81 30017171-3 2018 The purpose of this research was to evaluate the effects of fluconazole, which is known to inhibit CYP2C9, on imrecoxib"s pharmacokinetic (PK) parameters. Fluconazole 60-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 30017171-3 2018 The purpose of this research was to evaluate the effects of fluconazole, which is known to inhibit CYP2C9, on imrecoxib"s pharmacokinetic (PK) parameters. Imrecoxib 110-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 30017171-14 2018 Hence, it is necessary to adjust the imrecoxib dose when it is concurrently used with other CYP2C9 inhibitors. Imrecoxib 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 30415235-5 2018 CYP2C9 and VKORC1 polymorphisms are essential determinants in the genotype-guided dosing of warfarin and may distinguish patients who would benefit from switching to direct oral anticoagulants (DOACs). Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30415235-5 2018 CYP2C9 and VKORC1 polymorphisms are essential determinants in the genotype-guided dosing of warfarin and may distinguish patients who would benefit from switching to direct oral anticoagulants (DOACs). doacs 194-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30065771-8 2018 It was indicated that low warfarin dose requirements are strongly associated with the presence of CYP2C9 and VKORC1-1639 variant alleles. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 29621561-0 2018 Different inhibitory effects of azole-containing drugs and pesticides on CYP2C9 polymorphic forms: An in vitro study. Azoles 32-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 29621561-4 2018 Due to the similarities in CYP organization throughout species, azoles can interact not only with the target fungal CYP51 substrate-binding site but can also modulate the catalytic activity of human cytochrome P450s, including CYP2C9, causing severe adverse effects. Azoles 64-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 227-233 29621561-5 2018 In the present study the potency of azole-containing drugs and pesticides to inhibit recombinant wild-type CYP2C9*1 and the allelic variants CYP2C9*2 and CYP2C9*3 was evaluated. Azoles 36-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 29621561-5 2018 In the present study the potency of azole-containing drugs and pesticides to inhibit recombinant wild-type CYP2C9*1 and the allelic variants CYP2C9*2 and CYP2C9*3 was evaluated. Azoles 36-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 29621561-5 2018 In the present study the potency of azole-containing drugs and pesticides to inhibit recombinant wild-type CYP2C9*1 and the allelic variants CYP2C9*2 and CYP2C9*3 was evaluated. Azoles 36-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 29621561-7 2018 Moreover, addition of cytochrome b5 resulted in a decrease of CYP2C9*3 activity to diclofenac in a concentration-dependent manner. Diclofenac 83-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 29621561-8 2018 Increasing the knowledge of how azoles influence polymorphic variants of CYP2C9 could help individualize drug treatment, leading to optimization of the selection of drugs and doses for individuals based on genetic information. Azoles 32-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 30065771-1 2018 Polymorphism in the genes encoding CYP2C9 enzyme and VKORC1 reductase significantly influence warfarin dose requirement since patients with CYP2C9*2, CYP2C9*3 and VKORC1 mutant alleles require lower warfarin maintenance doses. Warfarin 199-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 30030468-0 2018 CYP2C9 and OATP1B1 genetic polymorphisms affect the metabolism and transport of glimepiride and gliclazide. glimepiride 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30030468-6 2018 The clearance of glimepiride and gliclazide in CYP2C9*2 and *3 was significantly reduced compared to the wild-type. glimepiride 17-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 30030468-0 2018 CYP2C9 and OATP1B1 genetic polymorphisms affect the metabolism and transport of glimepiride and gliclazide. Gliclazide 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30030468-6 2018 The clearance of glimepiride and gliclazide in CYP2C9*2 and *3 was significantly reduced compared to the wild-type. Gliclazide 33-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 30030468-2 2018 The aim of this study was to assess the effect of CYP2C9 and OATP1B1 genetic polymorphisms on the metabolism and transport of glimepiride and gliclazide. glimepiride 126-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 30030468-7 2018 These findings collectively indicate that OATP1B1*5 and *15 and CYP2C9*2 and *3 have a significant effect on the transport and metabolism of glimepiride and gliclazide. glimepiride 141-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 30030468-2 2018 The aim of this study was to assess the effect of CYP2C9 and OATP1B1 genetic polymorphisms on the metabolism and transport of glimepiride and gliclazide. Gliclazide 142-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 30030468-7 2018 These findings collectively indicate that OATP1B1*5 and *15 and CYP2C9*2 and *3 have a significant effect on the transport and metabolism of glimepiride and gliclazide. Gliclazide 157-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 30030468-5 2018 Glimepiride in the presence of CYP2C9*1, *2 and *3 recombinase had Vmax values of 21.58 +- 7.78, 15.69 +- 5.59, and 9.17 +- 3.03 nmol/min/mg protein, while gliclazide had Vmax values of 15.73 +- 3.11, 10.53 +- 4.06, and 6.21 +- 2.94 nmol/min/mg protein, respectively. glimepiride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 30030468-5 2018 Glimepiride in the presence of CYP2C9*1, *2 and *3 recombinase had Vmax values of 21.58 +- 7.78, 15.69 +- 5.59, and 9.17 +- 3.03 nmol/min/mg protein, while gliclazide had Vmax values of 15.73 +- 3.11, 10.53 +- 4.06, and 6.21 +- 2.94 nmol/min/mg protein, respectively. Gliclazide 156-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 29781049-1 2018 PURPOSE: This study aimed to investigate relationships between times in therapeutic range (TTR) or warfarin sensitivity indexes (WSI) and VKORC1-1639G>A and CYP2C9 polymorphisms in patients with left ventricular assist devices (LVAD). Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 30034221-0 2018 Does celecoxib inhibit agomelatine metabolism via CYP2C9 or CYP1A2? Celecoxib 5-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 29377603-11 2018 The changes in the mean of the Cmax and AUCtau of celecoxib were greater in intermediate metabolizers of cytochrome 2C9 (CYP2C9) than in normal metabolizers. Celecoxib 50-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 29794812-1 2018 : Mutations in the genes encoding vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) largely contribute to the inter-individual variations in vitamin K antagonists (VKAs) dose requirements. Vitamin K 34-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 29628024-0 2018 The association between CYP2C9/2C19 polymorphisms and phenytoin maintenance doses in Asian epileptic patients: A systematic review and meta-analysis : . Phenytoin 54-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 29628024-5 2018 RevMan 5.2.3 software was used to analyze the relationship between CYP2C9/2C19 polymorphisms and PHT maintenance doses. Phenytoin 97-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 29628024-9 2018 Patients with heterozygous CYP2C9 or both heterozygous CYP2C9 and CYP2C19 showed a trend but not a statistically-significant decrease of PHT dose, but dosage adjustment was recommended. Phenytoin 137-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 29628024-9 2018 Patients with heterozygous CYP2C9 or both heterozygous CYP2C9 and CYP2C19 showed a trend but not a statistically-significant decrease of PHT dose, but dosage adjustment was recommended. Phenytoin 137-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29628024-10 2018 CONCLUSION: The meta-analysis indicates that CYP2C9 and CYP2C19 polymorphisms are associated with lower PHT maintenance dosage in Asian epileptic patients. Phenytoin 104-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 29789925-0 2018 Associations of polymorphisms of CYP2D6 and CYP2C9 with early onset severe pre-eclampsia and response to labetalol therapy. Labetalol 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 29789925-3 2018 The aim of this study was to evaluate the associations of polymorphisms of cytochrome P450, family 2, subfamily D, polypeptide 6 (CYP2D6) and cytochrome P450, family 2, subfamily C, polypeptide 9 (CYP2C9) with PPE and the relationship among CYP2D6, CYP2C9 polymorphisms and response to labetalol therapy. Labetalol 286-295 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-195 29989011-7 2018 However, at 10 microM TQ, CYP2C9 enzyme activity was maximally inhibited by 69.69%, followed by CYP3A4 (23.59%) > CYP1A2 (23.51%) > CYP2D6 (11.42%). thymoquinone 22-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 29578577-2 2018 Posaconazole (POS), a substrate of glucuronosyltransferase and P-glycoprotein, and voriconazole (VRC), a substrate of CYP2C9/19, are commonly administered to transplant recipients. Voriconazole 83-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 29578577-2 2018 Posaconazole (POS), a substrate of glucuronosyltransferase and P-glycoprotein, and voriconazole (VRC), a substrate of CYP2C9/19, are commonly administered to transplant recipients. Voriconazole 97-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 29578577-13 2018 These results suggest that letermovir may be a perpetrator of CYP2C9/19-mediated drug-drug interactions. letermovir 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 29989011-1 2018 The aim of the present study was to investigate the potential effect of thymoquinone (TQ) on the metabolic activity of four major drug metabolizing enzymes in human liver microsomes, namely cytochrome P450 (CYP) 1A2, CYP2C9, CYP2D6 and CYP3A4. thymoquinone 72-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 29989011-1 2018 The aim of the present study was to investigate the potential effect of thymoquinone (TQ) on the metabolic activity of four major drug metabolizing enzymes in human liver microsomes, namely cytochrome P450 (CYP) 1A2, CYP2C9, CYP2D6 and CYP3A4. thymoquinone 86-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 29989011-2 2018 The inhibition of CYP enzymatic activities by TQ was evaluated by incubating typical substrates (phenacetin for CYP1A2, tolbutamide for CYP2C9, dextromethorphan for CYP2D6, and testosterone for CYP3A4) with human liver microsomes and NADPH in the absence or presence of TQ (1, 10 and 100 microM). thymoquinone 46-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 29778033-3 2018 Polymorphisms in the vitamin K expoxide reductase 1 (VKORC1) and cytochrome P4502C9 (CYP2C9) genes have been reported to influence the warfarin dose. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 29778033-4 2018 We investigated the effects of the VKORC1 and CYP2C9 genotypes on the warfarin dose in pediatric patients with giant CAAs after KD. Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 29778033-11 2018 Three patients with CYP2C9*1/*3 had a lower warfarin dose than did those with the wild CYP2C9*1/*1 genotype, but the difference did not reach significance (p > 0.05). Warfarin 44-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 29119333-5 2018 METHODS: A PBPK model was optimized in SimCYP for tolbutamide as a CYP2C9 substrate, based on published in vitro and clinical data. Tolbutamide 51-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 29620050-13 2018 Based on early reported human pharmacokinetics, drug interaction indices were 0.16 for CYP3A4 and 0.02 for CYP2C9, suggesting that anlotinib had a low propensity to precipitate drug interactions on these enzymes. anlotinib 131-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 29119333-0 2018 Physiologically Based Pharmacokinetic Modelling of Cytochrome P450 2C9-Related Tolbutamide Drug Interactions with Sulfaphenazole and Tasisulam. Tolbutamide 79-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-70 28737453-16 2018 Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. selexipag 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 28737453-16 2018 Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. selexipag 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 28520347-0 2012 Warfarin Therapy and the Genotypes CYP2C9 and VKORC1 Warfarin is an anticoagulant that acts by reducing the activity of vitamin K-dependent clotting factors. Vitamin K 120-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 28520347-3 2012 A patient"s CYP2C9 and VKORC1 genotype can be used to help determine the optimal starting dose of warfarin. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 28520347-4 2012 The CYP2C9 gene encodes one of the main enzymes involved in the metabolism of warfarin. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 28520347-5 2012 Several variant CYP2C9 alleles are associated with reduced enzyme activity and lower clearance rates of warfarin. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 28520347-6 2012 Patients who carry at least one copy of such a variant allele (such as CYP2C9*2 and CYP2C9*3) have reduced metabolism leading to higher warfarin concentrations. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 28520347-6 2012 Patients who carry at least one copy of such a variant allele (such as CYP2C9*2 and CYP2C9*3) have reduced metabolism leading to higher warfarin concentrations. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 28520347-7 2012 On average, they require a lower daily warfarin dose than patients who are homozygous for the wild-type CYP2C9*1 allele. Warfarin 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28520347-10 2012 The FDA-approved warfarin drug label provides a dosing table based on CYP2C9 and VKORC1 genotypes (Table 1). Warfarin 17-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29710546-0 2018 Pharmacokinetic interaction of diosmetin and silibinin with other drugs: Inhibition of CYP2C9-mediated biotransformation and displacement from serum albumin. diosmetin 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 29710546-0 2018 Pharmacokinetic interaction of diosmetin and silibinin with other drugs: Inhibition of CYP2C9-mediated biotransformation and displacement from serum albumin. Silybin 45-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 29710546-5 2018 In this study, the inhibitory action of DIO and SIL on CYP2C9-catalyzed metabolism of diclofenac to 4"-hydroxydiclofenac was examined, using warfarin as a positive control. diosmetin 40-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29710546-5 2018 In this study, the inhibitory action of DIO and SIL on CYP2C9-catalyzed metabolism of diclofenac to 4"-hydroxydiclofenac was examined, using warfarin as a positive control. Diclofenac 86-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29710546-5 2018 In this study, the inhibitory action of DIO and SIL on CYP2C9-catalyzed metabolism of diclofenac to 4"-hydroxydiclofenac was examined, using warfarin as a positive control. 4'-hydroxydiclofenac 100-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29710546-5 2018 In this study, the inhibitory action of DIO and SIL on CYP2C9-catalyzed metabolism of diclofenac to 4"-hydroxydiclofenac was examined, using warfarin as a positive control. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29710546-7 2018 It is demonstrated that DIO and SIL are potent inhibitors of CYP2C9 enzyme and are able to displace the Site I ligand warfarin from human serum albumin. diosmetin 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29710546-7 2018 It is demonstrated that DIO and SIL are potent inhibitors of CYP2C9 enzyme and are able to displace the Site I ligand warfarin from human serum albumin. Silybin 32-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29710546-7 2018 It is demonstrated that DIO and SIL are potent inhibitors of CYP2C9 enzyme and are able to displace the Site I ligand warfarin from human serum albumin. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29119333-0 2018 Physiologically Based Pharmacokinetic Modelling of Cytochrome P450 2C9-Related Tolbutamide Drug Interactions with Sulfaphenazole and Tasisulam. Sulfaphenazole 114-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-70 29119333-0 2018 Physiologically Based Pharmacokinetic Modelling of Cytochrome P450 2C9-Related Tolbutamide Drug Interactions with Sulfaphenazole and Tasisulam. N-((5-bromo-2-thienyl)sulfonyl)-2,4-dichlorobenzamide 133-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-70 29119333-11 2018 CONCLUSIONS: This optimized tolbutamide PBPK model was verified with two strong CYP2C9 inhibitors and can be applied to the prediction of CYP2C9 interactions for novel inhibitors. Tolbutamide 28-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 29119333-2 2018 Previously published physiologically based pharmacokinetic (PBPK) models of tolbutamide are based on an assumption that its metabolic clearance is exclusively through CYP2C9; however, many studies indicate that CYP2C9 metabolism is only responsible for 80-90% of the total clearance. Tolbutamide 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 29119333-11 2018 CONCLUSIONS: This optimized tolbutamide PBPK model was verified with two strong CYP2C9 inhibitors and can be applied to the prediction of CYP2C9 interactions for novel inhibitors. Tolbutamide 28-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 29119333-2 2018 Previously published physiologically based pharmacokinetic (PBPK) models of tolbutamide are based on an assumption that its metabolic clearance is exclusively through CYP2C9; however, many studies indicate that CYP2C9 metabolism is only responsible for 80-90% of the total clearance. Tolbutamide 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 211-217 29746595-2 2018 The allelic variant CYP2C9*30 (A477T) is associated to diminished response to the antihypertensive effects of the prodrug losartan and affected metabolism of other drugs. Losartan 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 29119333-4 2018 This paper describes the development and verification of SimCYP -based PBPK models that accurately describe the human pharmacokinetics of tolbutamide when dosed alone or in combination with the CYP2C9 inhibitors sulfaphenazole and tasisulam. Tolbutamide 138-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 29274302-0 2018 Association of CYP2C9*3 with phenytoin-induced Stevens-Johnson syndrome and toxic epidermal necrolysis: A systematic review and meta-analysis. Phenytoin 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 29274302-3 2018 The aim of this meta-analysis was to evaluate the association between CYP2C9*3 and PHT-induced SJS/TEN. Phenytoin 83-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29274302-5 2018 Studies exploring the relationship between CYP2C9*3 and PHT-induced SJS and TEN were included. Phenytoin 56-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 29274302-10 2018 WHAT IS NEW AND CONCLUSION: A significant association between CYP2C9*3 and PHT-induced SJS/TEN was identified, especially in a Thai population. Phenytoin 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 29274302-11 2018 CYP2C9*3 is thus a credible predictive genetic marker of PHT-induced SJS/TEN. Phenytoin 57-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29530595-6 2018 Here, pulsed and continuous wave (CW) EPR demonstrate that water-bridged complexes are remarkably common among a range of nitrogenous drugs or drug fragments that bind to CYP3A4 or CYP2C9. Water 59-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 29880089-1 2018 In chronic kidney disease (CKD) patients, the ratio of warfarin enantiomers is changed and becomes unstable due to a reduction of cytochrome P450 (CYP) 2C9 activity of, which contributes to the development of hemorrhagic complications. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-155 29538081-8 2018 RECENT FINDINGS: DAH has been associated with the presence of variant alleles in vitamin K epoxide reductase complex 1, cytochrome P450 (CYP)2C9 and CYP2C19 genes. dah 17-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-144 29475833-1 2018 Bosentan is a substrate of hepatic uptake transporter organic anion-transporting polypeptides (OATPs), and undergoes extensive hepatic metabolism by cytochrome P450 (P450), namely, CYP3A4 and CYP2C9. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 29368187-1 2018 PURPOSE: The objective of this study is to assess the effects of green tea and its major catechin component, (-)-epigallocatechin gallate (EGCG), on CYP2C9-mediated substrate metabolism in vitro, and the pharmacokinetics of fluvastatin in healthy volunteers. Catechin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 28401802-12 2018 This study found that routine medication with warfarin provides significantly different levels of anticoagulant control between patients with wild-type genotypes and patients with heterozygous polymorphism genotypes of VKORC1 rs9923231 or CYP2C9 rs1057910. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 239-245 28401802-13 2018 Patients with heterozygous polymorphism genotypes of VKORC1 or CYP2C9 require genotype-directed therapy with warfarin to increase efficacy and safety in anticoagulant treatment. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 28401802-1 2018 Differences in warfarin maintenance dosages based on the presence of polymorphisms in VKORC1, CYP2C9, CYP4F2, and ORM1 can be determined through dosage adjustment according to routine guidelines. Warfarin 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 29368187-1 2018 PURPOSE: The objective of this study is to assess the effects of green tea and its major catechin component, (-)-epigallocatechin gallate (EGCG), on CYP2C9-mediated substrate metabolism in vitro, and the pharmacokinetics of fluvastatin in healthy volunteers. epigallocatechin gallate 109-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 29368187-1 2018 PURPOSE: The objective of this study is to assess the effects of green tea and its major catechin component, (-)-epigallocatechin gallate (EGCG), on CYP2C9-mediated substrate metabolism in vitro, and the pharmacokinetics of fluvastatin in healthy volunteers. epigallocatechin gallate 139-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 29368187-2 2018 METHODS: The metabolism of diclofenac and fluvastatin in human recombinant CYP2C9 was investigated in the presence of EGCG. Diclofenac 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 29368187-2 2018 METHODS: The metabolism of diclofenac and fluvastatin in human recombinant CYP2C9 was investigated in the presence of EGCG. Fluvastatin 42-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 29368187-2 2018 METHODS: The metabolism of diclofenac and fluvastatin in human recombinant CYP2C9 was investigated in the presence of EGCG. epigallocatechin gallate 118-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 29368187-9 2018 CONCLUSIONS: Although in vitro studies indicated that EGCG and brewed green tea produce significant inhibitory effects on CYP2C9 activity, the concomitant administration of green tea and fluvastatin in healthy volunteers did not influence the pharmacokinetics of fluvastatin. epigallocatechin gallate 54-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 29596975-4 2018 CYP2C9, CYP3A4, and CYP3A5 were involved in the formation of FMS S-oxide, which was further metabolized to BR-A-557 by CYP3A4/5. Oxides 65-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29596975-5 2018 CYP2C9 played an exclusive role in n-butyl hydroxylation. Nitrogen 15-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29596975-9 2018 In addition, simultaneous determination of the FMS metabolites may be used to evaluate CYP2C9 and CYP3A4/5 activity. fimasartan 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 29522717-0 2018 Altered metabolism of synthetic cannabinoid JWH-018 by human cytochrome P450 2C9 and variants. 1-pentyl-3-(1-naphthoyl)indole 44-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-80 29724296-2 2018 METHOD: A total of 22 patients who required an extreme warfarin dosage from VKORC1 -1639AA & CYP2C9*1*1 genotype group were enrolled in this study. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 29522717-3 2018 The major enzyme involved in the metabolism of JWH-018 is CYP2C9, a highly polymorphic enzyme found largely in the intestines and liver, with *1 being designated as the wild type, and *2 and *3 as the two most common variants. (4-methyl-1-naphthyl)-(1-pentylindol-3-yl)methanone 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 29522717-6 2018 Here, in vitro enzyme kinetic assays performed with human recombinant CYP2C9 variants (*1, *2, and *3) revealed that oxidative metabolism by CYP2C9*3 resulted in significantly less formation of (omega)-OH and (omega-1)-OH metabolites. (omega)-oh 194-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29522717-6 2018 Here, in vitro enzyme kinetic assays performed with human recombinant CYP2C9 variants (*1, *2, and *3) revealed that oxidative metabolism by CYP2C9*3 resulted in significantly less formation of (omega)-OH and (omega-1)-OH metabolites. (omega)-oh 194-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 29522717-6 2018 Here, in vitro enzyme kinetic assays performed with human recombinant CYP2C9 variants (*1, *2, and *3) revealed that oxidative metabolism by CYP2C9*3 resulted in significantly less formation of (omega)-OH and (omega-1)-OH metabolites. (omega-1)-oh 209-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29522717-6 2018 Here, in vitro enzyme kinetic assays performed with human recombinant CYP2C9 variants (*1, *2, and *3) revealed that oxidative metabolism by CYP2C9*3 resulted in significantly less formation of (omega)-OH and (omega-1)-OH metabolites. (omega-1)-oh 209-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 29522717-7 2018 Surprisingly, CYP2C9*2 was roughly 3.6-fold more efficient as the CYP2C9*1 enzyme based on Vmax/Km, increasing the rate of JWH-018 metabolism and allowed for a much more rapid elimination. (4-methyl-1-naphthyl)-(1-pentylindol-3-yl)methanone 123-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 29522717-7 2018 Surprisingly, CYP2C9*2 was roughly 3.6-fold more efficient as the CYP2C9*1 enzyme based on Vmax/Km, increasing the rate of JWH-018 metabolism and allowed for a much more rapid elimination. (4-methyl-1-naphthyl)-(1-pentylindol-3-yl)methanone 123-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 29622878-0 2018 The Influence of CYP2C9 and VKORC1 Gene Polymorphisms on the Response to Warfarin in Egyptians. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 29488228-4 2018 Utilizing valsartan hydroxylase assay as probe, all forms of glucosamine and chondroitin exhibited IC50 values beyond 1000 muM, indicating very weak potential in inhibiting CYP2C9. Glucosamine 61-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 29488228-4 2018 Utilizing valsartan hydroxylase assay as probe, all forms of glucosamine and chondroitin exhibited IC50 values beyond 1000 muM, indicating very weak potential in inhibiting CYP2C9. Chondroitin 77-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 29273968-3 2018 SimCYP, a physiologically based PK model, was developed and used to predict the effects of fluconazole and CYP2C9 genetic polymorphism on siponimod metabolism. siponimod 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 29273968-5 2018 RESULTS: Correlation analysis suggested that CYP2C9 is the main enzyme responsible for siponimod metabolism in humans. siponimod 87-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 29273968-6 2018 Compared with the CYP2C9*1/*1 genotype, HLM incubations from CYP2C9*3/*3 and CYP2C9*2/*2 donors showed ~ 10- and 3-fold decrease in siponimod metabolism, respectively. siponimod 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29273968-6 2018 Compared with the CYP2C9*1/*1 genotype, HLM incubations from CYP2C9*3/*3 and CYP2C9*2/*2 donors showed ~ 10- and 3-fold decrease in siponimod metabolism, respectively. siponimod 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 29273968-7 2018 Simulations of enzyme contribution predicted that in the CYP2C9*1/*1 genotype, CYP2C9 is predominantly responsible for siponimod metabolism (~ 81%), whereas in the CYP2C9*3/*3 genotype, its contribution is reduced to 11%. siponimod 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 29273968-7 2018 Simulations of enzyme contribution predicted that in the CYP2C9*1/*1 genotype, CYP2C9 is predominantly responsible for siponimod metabolism (~ 81%), whereas in the CYP2C9*3/*3 genotype, its contribution is reduced to 11%. siponimod 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 29273968-7 2018 Simulations of enzyme contribution predicted that in the CYP2C9*1/*1 genotype, CYP2C9 is predominantly responsible for siponimod metabolism (~ 81%), whereas in the CYP2C9*3/*3 genotype, its contribution is reduced to 11%. siponimod 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 29273968-8 2018 The predicted exposure increase of siponimod with fluconazole 200 mg was 2.0-2.4-fold for CYP2C9*1/*1 genotype. siponimod 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 29622878-3 2018 Cytochrome P-450 2C9 is the principle enzyme that terminates the anticoagulant effect of warfarin by catalyzing the conversion of the pharmacologically more potent S-enantiomer to its inactive metabolites. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-20 29273968-8 2018 The predicted exposure increase of siponimod with fluconazole 200 mg was 2.0-2.4-fold for CYP2C9*1/*1 genotype. Fluconazole 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 29273968-12 2018 In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 29622878-6 2018 The current study aimed to investigate the pharmacogenetic effect of CYP2C9 and VKORC1 gene polymorphisms on the patients response to warfarin. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 29273968-12 2018 In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 29622878-19 2018 Data from our study suggest that together with clinical factors, VKORC1 and CYP2C9 polymorphisms are important contributors to warfarin dosing and may help predict adverse effects in Egyptian patients. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 29273968-12 2018 In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 29273968-12 2018 In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes. siponimod 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 29623958-0 2018 Correction to: The Influence of CYP2C9 and VKORC1 Gene Polymorphisms on the Response to Warfarin in Egyptians. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 29563776-4 2018 In an in vitro study, celecoxib with a series of concentrations was added to an incubation mixture containing recombinant human CYP2C9, human or rat liver microsomes to determine the half-maximal inhibitory concentration on the metabolism of agomelatine. Celecoxib 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 29636628-0 2018 CYP2C9 polymorphisms in epilepsy: influence on phenytoin treatment. Phenytoin 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29636628-3 2018 Genetic polymorphism of CYP2C9 may reduce the metabolism of PHT by 25-50% in patients with variants *2 and *3 compared to those with wild-type variant *1. Phenytoin 60-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 29636628-7 2018 The presence of HLA-B*15:02 and CYP2C9 *2 or *3 in the same patient increases the risk of Stevens-Johnson syndrome and toxic epidermal necrolysis; hence, PHT should not be prescribed in these patients. Phenytoin 154-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 29636628-8 2018 In patients with CYP2C9 *1/*2 or *1/*3 alleles (intermediate metabolizers), the usual PHT maintenance dose (5-10 mg/kg/day) must be reduced by 25%, and in those with CYP2C9 *2/*2, *2/*3 or *3/*3 alleles (poor metabolizers), the dose must be reduced by 50%. Phenytoin 86-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 29636628-10 2018 In this paper, we aim to review the influence of CYP2C9 polymorphism on the metabolization of PHT and the clinical implications of poor metabolization in the treatment of epilepsies. Phenytoin 94-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 29563776-5 2018 Moreover, a mechanism study was performed to determine the inhibitory effect of celecoxib on CYP2C9. Celecoxib 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 29563776-7 2018 In addition, celecoxib inhibited the metabolism of agomelatine in the in vitro studies, which was determined to be by a competitive mechanism on CYP2C9. Celecoxib 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 29563776-7 2018 In addition, celecoxib inhibited the metabolism of agomelatine in the in vitro studies, which was determined to be by a competitive mechanism on CYP2C9. agomelatine 51-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 29433307-2 2018 Cytochrome P450 2C9 ( CYP2C9) genotype-guided warfarin dosing is recommended in the clinic, given the more potent S-warfarin is primarily metabolized by CYP2C9. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 29433307-3 2018 However, only about 20-30% of interpatient variability in S-warfarin clearance is associated with CYP2C9 genotype. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 29105855-0 2018 Dicloxacillin induces CYP2C19, CYP2C9 and CYP3A4 in vivo and in vitro. Dicloxacillin 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 27879469-11 2018 Although Cytochrome P450 2C9 (CYP2C9) and VKORC1 polymorphisms together attributed only 3.8% variability in warfarin dose but it was statistically significant ( p value = .004). Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-28 29353070-2 2018 Cytochrome P450 2C9 (CYP2C9) plays the key role in transformation of coumarin and thus, influences determination of warfarin dosage. coumarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 29353070-2 2018 Cytochrome P450 2C9 (CYP2C9) plays the key role in transformation of coumarin and thus, influences determination of warfarin dosage. coumarin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 29353070-2 2018 Cytochrome P450 2C9 (CYP2C9) plays the key role in transformation of coumarin and thus, influences determination of warfarin dosage. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 29353070-2 2018 Cytochrome P450 2C9 (CYP2C9) plays the key role in transformation of coumarin and thus, influences determination of warfarin dosage. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 29353070-3 2018 A number of factors including dietary compounds such as sesamin, caffeic acid and ferulic acids can regulate the activity of CYP2C9. sesamin 56-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 29353070-3 2018 A number of factors including dietary compounds such as sesamin, caffeic acid and ferulic acids can regulate the activity of CYP2C9. caffeic acid 65-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 29353070-3 2018 A number of factors including dietary compounds such as sesamin, caffeic acid and ferulic acids can regulate the activity of CYP2C9. ferulic acid 82-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 29353070-4 2018 The present study tested the hypothesis that sesamin, episesamin, caffeic acid and ferulic acid decreases the rate of warfarin 7-hydroxylation via inhibition of hepatic CYP2C9. sesamin 45-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 29353070-4 2018 The present study tested the hypothesis that sesamin, episesamin, caffeic acid and ferulic acid decreases the rate of warfarin 7-hydroxylation via inhibition of hepatic CYP2C9. sesamin 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 27879469-11 2018 Although Cytochrome P450 2C9 (CYP2C9) and VKORC1 polymorphisms together attributed only 3.8% variability in warfarin dose but it was statistically significant ( p value = .004). Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 29353070-4 2018 The present study tested the hypothesis that sesamin, episesamin, caffeic acid and ferulic acid decreases the rate of warfarin 7-hydroxylation via inhibition of hepatic CYP2C9. caffeic acid 66-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 28049362-5 2018 Five variables, that is, VKORC1-1639 G>A, CYP2C9*2, CYP2C9*3, age, and diet, were found to be significantly associated with warfarin response in univariate analysis. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 29353070-4 2018 The present study tested the hypothesis that sesamin, episesamin, caffeic acid and ferulic acid decreases the rate of warfarin 7-hydroxylation via inhibition of hepatic CYP2C9. ferulic acid 83-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 28049362-5 2018 Five variables, that is, VKORC1-1639 G>A, CYP2C9*2, CYP2C9*3, age, and diet, were found to be significantly associated with warfarin response in univariate analysis. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 29404794-3 2018 RESULTS: Investigated steroids usually reveal a high gastrointestinal absorption and a good oral bioavailability, may inhibit someof the human cytochromes involved in the metabolism of xenobiotics (CYP2C9 being the most affected) and reflect a good capacity for skin penetration. Steroids 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 29326367-0 2018 Organic Anion Transporter 2 Mediates Hepatic Uptake of Tolbutamide, a CYP2C9 Probe Drug. Tolbutamide 55-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 29326367-1 2018 Tolbutamide is primarily metabolized by CYP2C9, and, thus, is frequently applied as a clinical probe substrate for CYP2C9 activity. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 29326367-1 2018 Tolbutamide is primarily metabolized by CYP2C9, and, thus, is frequently applied as a clinical probe substrate for CYP2C9 activity. Tolbutamide 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 29326367-9 2018 In contrast to the rapid equilibrium (CYP2C9-only) model, the permeability-limited (OAT2-CYP2C9 interplay) model better described the plasma concentration-time profiles of tolbutamide. Tolbutamide 172-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 29326367-10 2018 Additionally, the latter well described tolbutamide pharmacokinetics in carriers of CYP2C9 genetic variants and quantitatively rationalized its known drug-drug interactions. Tolbutamide 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 29568565-1 2018 Aim: To evaluate applicability of CYP2C9*2, *3 and VKORC1-1639G > A based algorithm to predict warfarin stable dose (WSD) in a group of Palestinian patients. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 29305929-5 2018 The production of 11-hydroxylauric acid was mediated by CYP2E1, CYP2C9, CYP2B6, CYP1A2, CYP3A4, and CYP4A11. 11-hydroxydodecanoic acid 18-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 29168292-0 2018 Evaluation of the effect of quercetin treatment on CYP2C9 enzyme activity of diclofenac in healthy human volunteers. Quercetin 28-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29168292-0 2018 Evaluation of the effect of quercetin treatment on CYP2C9 enzyme activity of diclofenac in healthy human volunteers. Diclofenac 77-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 29168292-9 2018 The results suggest that quercetin might have inhibited CYP2C9-mediated metabolism of DIC. Quercetin 25-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 29314710-8 2018 Second, enzyme kinetic studies showed that the initial metabolic steps were formed by cytochrome P450 isoforms (CYP) CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 resulting in pyrrolidine, thiophene or alkyl hydroxy metabolites depending on the length of the alkyl chain. pyrrolidine 182-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 29314710-8 2018 Second, enzyme kinetic studies showed that the initial metabolic steps were formed by cytochrome P450 isoforms (CYP) CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 resulting in pyrrolidine, thiophene or alkyl hydroxy metabolites depending on the length of the alkyl chain. Thiophenes 195-204 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 30135636-10 2018 CYP2C9 is a major enzyme responsible for the metabolism of phenytoin. Phenytoin 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28656666-1 2018 Data on the association of CYP2C9 genetic polymorphisms with sulfonylurea (SU)-induced hypoglycaemia (SH) are inconsistent. Sulfonylurea Compounds 61-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 29542427-9 2018 Meanwhile, all individual biomarkers showed no, less or moderate inhibitory effect towards all the tested CYP450 except for curcumin that showed inhibition of CYP2C8 (91%), CYP2C9 (81%) and CYP2C19 (72%) at 10microM. Curcumin 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 173-179 29542427-10 2018 CONCLUSION: Curcumin was found to be an active constituent that might contribute to the inhibition of SynacinnTM against CYP2C8, CYP2C9 and CYP2C19. Curcumin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 29760340-5 2018 The age and SNPs in VKORC1 and CYP2C9 were significantly associated with the weekly acenocoumarol dose requirement (p = 0.023, p = 0.0001 and p = 0.001 respectively). Acenocoumarol 84-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 29760340-7 2018 Non-parametric analysis confirmed the accumulate effect of variant alleles at VKORC1 -1639 G>A, VKORC1 1173 C>T and CYP2C9 SNPs on the acenocoumarol dose requirement. Acenocoumarol 141-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 29760340-9 2018 The multiple linear regression analysis showed that mutation in VKORC1 -1639, VKORC1 1173 SNPs, or in CYP2C9 haplotype reduces the mean acenocoumarol weekly dose to 25.4%, 23.4% and 6.2%, respectively. Acenocoumarol 136-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. Triclabendazole 24-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. triclabendazole sulfoxide 41-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. Triclabendazole sulfone 68-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. montelukast 93-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. Montelukast sulfoxide 106-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 30117405-8 2018 CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 microM, 1.95 microM, 0.69 microM, 1.34 microM, 3.61 microM and 2.15 microM, respectively. Montelukast Sulfone 132-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28656666-1 2018 Data on the association of CYP2C9 genetic polymorphisms with sulfonylurea (SU)-induced hypoglycaemia (SH) are inconsistent. Sulfonylurea Compounds 75-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 28656666-6 2018 The number of CYP2C9 deficient alleles was associated with nearly three-fold higher odds of hypoglycaemia (OR, 2.81; 95% CI, 1.30-6.09; P = .009) and better response to SU treatment (beta, -0.218; SE, 0.074; P = .003) only in patients carrying the POR*1/*1 genotype. Sulfonylurea Compounds 169-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 28656666-7 2018 Our results indicate that interaction between CYP2C9 and POR genes may be an important determinant of efficacy and severe adverse effects of SU treatment. Sulfonylurea Compounds 141-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 27958378-4 2018 In ancestry-specific meta-analyses, eight novel pharmacogenomic loci met the threshold for genome-wide significance (P<5 x 10-8), and a pharmacokinetic variant in CYP2C9 (rs1057910) that has been associated with sulfonylurea-related treatment effects and other adverse drug reactions in previous studies was replicated. Sulfonylurea Compounds 215-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 29182754-1 2017 Many lines of evidence suggest that warfarin dosing variability is significantly associated with cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) variant alleles. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-116 29283396-6 2017 Warfarin is the most well studied CYP2C9 substrate; recent work on use of dosing algorithms that include CYP2C9 genotype to improve patient safety during initiation of warfarin dosing are reviewed and prospects for their clinical implementation considered. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 29283396-6 2017 Warfarin is the most well studied CYP2C9 substrate; recent work on use of dosing algorithms that include CYP2C9 genotype to improve patient safety during initiation of warfarin dosing are reviewed and prospects for their clinical implementation considered. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 29283396-6 2017 Warfarin is the most well studied CYP2C9 substrate; recent work on use of dosing algorithms that include CYP2C9 genotype to improve patient safety during initiation of warfarin dosing are reviewed and prospects for their clinical implementation considered. Warfarin 168-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 29252193-1 2017 BACKGROUND: We have earlier developed a pharmacogenomic algorithm for acenocoumarol dose prediction in Greek patients that included CYP2C9/VKORC1 genetic information. Acenocoumarol 70-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 29393278-5 2018 In general, ordonin has induced effects on the major member of CYP450s mRNA and protein expression, as well as on the enzyme activity in human HepaRG cells, especially on CYP3A4 and CYP2C9. ordonin 12-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 29503429-5 2018 Whereas sesamin metabolism is mainly mediated by CYP2C9 in human liver, sesamin causes a mechanism-based inhibition (MBI) of CYP2C9. sesamin 72-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 29503429-6 2018 However, we found that the metabolite-intermediate complex between CYP2C9 and sesamin was unstable, and the effects of sesamin appeared to be minimal. sesamin 78-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 29252193-7 2017 For CYP4F2, significant effects were found on patients" ability to reach stable dose and on acenocoumarol dose requirements when CYP2C9/VKORC1 sub-phenotypes were analyzed. Acenocoumarol 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 29252193-9 2017 Additionally, in CYP2C9 intermediate metabolizers (IMs), after adjusting for age, weight, and VKORC1 genotypes, CYP4F2 genotypes were significantly associated with acenocoumarol stable dose (beta: 0.07; 95% CI: 0.006-0.134; p=0.033). Acenocoumarol 164-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 29182754-1 2017 Many lines of evidence suggest that warfarin dosing variability is significantly associated with cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) variant alleles. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 29182754-5 2017 We also found that patients with CYP2C9 *1 and VKORC1 -1639G alleles required the highest dosages of warfarin, while the carriers of CYP2C9 variant *2 and *3 alleles and VKORC1 -1639A required less warfarin. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 29182754-5 2017 We also found that patients with CYP2C9 *1 and VKORC1 -1639G alleles required the highest dosages of warfarin, while the carriers of CYP2C9 variant *2 and *3 alleles and VKORC1 -1639A required less warfarin. Warfarin 198-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 29182754-5 2017 We also found that patients with CYP2C9 *1 and VKORC1 -1639G alleles required the highest dosages of warfarin, while the carriers of CYP2C9 variant *2 and *3 alleles and VKORC1 -1639A required less warfarin. Warfarin 198-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 29182754-6 2017 Univariate regression analysis showed that age and presence of CYP2C9 *2 allele significantly influenced the daily warfarin dose requirement. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 29182754-7 2017 Our findings provide additional evidence to support the hypothesis that CYP2C9*2 (430C/T), *3 (1075A/C) and VKORC1 (-1639G/A) polymorphisms explain a considerable proportion of interindividual variability in warfarin dose. Warfarin 208-216 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 29148818-0 2017 Dissociation Constants of Cytochrome P450 2C9/Cytochrome P450 Reductase Complexes in a Lipid Bilayer Membrane Depend on NADPH: A Single-Protein Tracking Study. NADP 120-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-45 29148818-7 2017 An Arrhenius analysis of diffusion constants was also carried out, demonstrating that the reduced forms of CPR and CYP2C9 interact differently with the biomimetic ER and may, in addition to protein conformational changes, contribute to the observed NADPH-dependent shift in Kd. NADP 249-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 28686288-2 2017 This study describes the differential distribution of amino acid substitution variants of CYP2C8 (*2-I269F & *3-R139K) and CYP2C9 (*2-C144R & *3-L359A) genes in 234 type 2 diabetes mellitus (T2DM) patients and 218 healthy controls from Andhra Pradesh, South India. Adenosine Monophosphate 145-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 29137842-8 2017 Extensive metabolism of cetrozole and TMD-322 was observed in the CYP2C19 expression system among the test CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). cetrozole 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 29055218-0 2017 CYP2C9 and VKORC1 in therapeutic dosing and safety of acenocoumarol treatment: implication for clinical practice in Hungary. Acenocoumarol 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29055218-1 2017 The purpose of this work was to investigate the contribution of CYP2C9 and VKORC1 to acenocoumarol (AC) dose variability, bleeding events in Hungary. Acenocoumarol 85-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 29055218-1 2017 The purpose of this work was to investigate the contribution of CYP2C9 and VKORC1 to acenocoumarol (AC) dose variability, bleeding events in Hungary. Acenocoumarol 100-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 29100760-2 2017 Human CYP2C9 exhibits extensive genetic polymorphism and numbers of clinic studies have demonstrated that CYP2C9 genetic polymorphism has a significant influence on the pharmacokinetics of benzbromarone. Benzbromarone 189-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 29024493-5 2017 Simulations using PK/PD models showed that CYP2C9 genotype is the factor that affects the PDs of meloxicam. 3-{1-[3-(Dimethylamino)propyl]-2-Methyl-1h-Indol-3-Yl}-4-(2-Methyl-1h-Indol-3-Yl)-1h-Pyrrole-2,5-Dione 90-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 29024493-5 2017 Simulations using PK/PD models showed that CYP2C9 genotype is the factor that affects the PDs of meloxicam. Meloxicam 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 29100760-2 2017 Human CYP2C9 exhibits extensive genetic polymorphism and numbers of clinic studies have demonstrated that CYP2C9 genetic polymorphism has a significant influence on the pharmacokinetics of benzbromarone. Benzbromarone 189-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 29100760-0 2017 Comparative study on the interaction between 3 CYP2C9 allelic isoforms and benzbromarone by using LC-MS/MS method. Benzbromarone 75-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 29100760-1 2017 Benzbromarone is a uricosuric drug metabolized predominantly by cytochrome P450 2C9 from in vitro findings. Benzbromarone 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-83 29100760-3 2017 But in vitro study on the interaction between CYP2C9 allelic isoforms and benzbromarone was rare. Benzbromarone 74-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 29100760-4 2017 Here, an LC-MS/MS method was established and validated to determine the concentration of benzbromarone in different CYP2C9 enzyme incubation systems for the drug-enzyme interaction study. Benzbromarone 89-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 29100760-6 2017 Based on this method, the interaction between 3 CYP2C9 allelic isoforms and benzbromarone was researched by kinetics parameters (Km, Vmax, Clint). Benzbromarone 76-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 29100760-7 2017 As a result, CYP2C9*1 displayed the highest metabolic activity towards benzbromarone, CYP2C9*2 showed a little lower catalytic activity than CYP2C9*1 (relative clearance/*1=85.86%), CYP2C9*3 showed the lowest catalytic activity (relative clearance/*1=21.57%). Benzbromarone 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 29100760-7 2017 As a result, CYP2C9*1 displayed the highest metabolic activity towards benzbromarone, CYP2C9*2 showed a little lower catalytic activity than CYP2C9*1 (relative clearance/*1=85.86%), CYP2C9*3 showed the lowest catalytic activity (relative clearance/*1=21.57%). Benzbromarone 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 29100760-7 2017 As a result, CYP2C9*1 displayed the highest metabolic activity towards benzbromarone, CYP2C9*2 showed a little lower catalytic activity than CYP2C9*1 (relative clearance/*1=85.86%), CYP2C9*3 showed the lowest catalytic activity (relative clearance/*1=21.57%). Benzbromarone 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 29100760-7 2017 As a result, CYP2C9*1 displayed the highest metabolic activity towards benzbromarone, CYP2C9*2 showed a little lower catalytic activity than CYP2C9*1 (relative clearance/*1=85.86%), CYP2C9*3 showed the lowest catalytic activity (relative clearance/*1=21.57%). Benzbromarone 71-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 29100760-8 2017 The result illustrated that various CYP2C9 allelic isoforms showed different enzymatic activities towards benzbromarone, which could offer effective consultation for personalized administration in clinic. Benzbromarone 106-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 27937048-3 2017 Polymorphisms of CYP3A4, CYP3A5, CYP2C9 and non-genetic factors such as age, gender, body mass index (BMI), transaminase levels, concomitant medications might also affect voriconazole initial steady serum trough concentration (VICmin) in haematological patients, but the effects were not clear. Voriconazole 171-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 28962866-5 2017 KEY FINDINGS: The in vitro metabolic enzyme of DCO was CYP3A4 (mediated the formation of metabolites M1-M5), CYP2C9 (mediated the formation of metabolites M1-M3, M6 and M8) and CYP2D6 (mediated the formation of metabolite M3) in HLM. 3,3-DICHLORO-2-PHOSPHONOMETHYL-ACRYLIC ACID 47-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 32095470-8 2017 Two SNPs (rs11165069 from ABCA4 (p=0.00002); rs17847036 from CYP2C9 (p=0.000001)) showed significant associations with talniflumate C max. talniflumate 119-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 28522373-1 2017 Carvedilol, a drug available as a racemic mixture, is metabolised into hydroxyphenylcarvedilol (OHC) by CYP2D6 and O-desmethylcarvedilol (DMC) by CYP2C9 followed by conjugation to glucuronides. Carvedilol 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 27414183-7 2017 The sulfate conjugation by SULT2A1 also contributed to the metabolism of TAK-438 to form TAK-438 N-sulfate, and CYP2C9 mediated the formation of M-IV-Sul from TAK-438 N-sulfate. tak-438 n-sulfate 159-176 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 28522373-1 2017 Carvedilol, a drug available as a racemic mixture, is metabolised into hydroxyphenylcarvedilol (OHC) by CYP2D6 and O-desmethylcarvedilol (DMC) by CYP2C9 followed by conjugation to glucuronides. hydroxyphenylcarvedilol 71-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 28522373-1 2017 Carvedilol, a drug available as a racemic mixture, is metabolised into hydroxyphenylcarvedilol (OHC) by CYP2D6 and O-desmethylcarvedilol (DMC) by CYP2C9 followed by conjugation to glucuronides. ohc 96-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 28522373-1 2017 Carvedilol, a drug available as a racemic mixture, is metabolised into hydroxyphenylcarvedilol (OHC) by CYP2D6 and O-desmethylcarvedilol (DMC) by CYP2C9 followed by conjugation to glucuronides. desmethylcarvedilol 138-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 28522373-7 2017 Even though data are limited, it appears that despite inhibition of CYP2C9 due to long-term glibenclamide administration to T2DM patients, overall no differences are observed in the total clearance of carvedilol when compared to healthy subjects (43.1 vs. 45.9L/h for (S)-(-)-carvedilol and 29.0 vs. 33.1L/h for (R)-(+)-carvedilol). Glyburide 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 28522373-8 2017 These results provide evidence of a compensatory mechanism for the inhibition of CYP2C9, with higher contribution of CYP2D6 activity to the elimination of carvedilol. Carvedilol 155-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 28689179-8 2017 CONCLUSION: In this study, CYP2C9 and VKORC1 genotypes identified patients with VTE at increased bleeding risk with warfarin. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 28941798-6 2017 The aim of this study was to investigate the potential inhibition of shikonin against CYP1A2, CYP2B1/6, CYP2C9/11, CYP2D1/6, CYP2E1 and CYP3A2/4 activities in human and rat liver microsomes through cocktail approach in vitro. shikonin 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28941798-8 2017 In human liver microsomes, shikonin was not only a mixed inhibitor of CYP1A2, CYP2B6, CYP2C9, CYP2D6 and CYP3A4, but also a competitive inhibitor of CYP2E1, with Ki values no more than 7.72muM. shikonin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 29099769-8 2017 R-AML was also a significantly more potent inhibitor of CYP2C9 (Ki 12.11 microM/S-AML 21.45 microM) and CYP2C19 (Ki 5.97 microM/S-AML 7.22 muM. Amlodipine 0-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 29076107-10 2017 CONCLUSION: These findings confirm that amenamevir (as a cytochrome P450 3A4 substrate) can interact with ketoconazole or rifampicin, and (as a cytochrome P450 3A4 inducer) can interact with midazolam; however, no interaction between amenamevir and (S)-warfarin was observed, indicating that amenamevir is not an inducer of cytochrome P450 2C9. ASP2151 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 324-343 28758225-0 2017 Metabolism of 7-ethoxycoumarin, flavanone and steroids by cytochrome P450 2C9 variants. 7-ethoxycoumarin 14-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-77 28758225-0 2017 Metabolism of 7-ethoxycoumarin, flavanone and steroids by cytochrome P450 2C9 variants. flavanone 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-77 28758225-0 2017 Metabolism of 7-ethoxycoumarin, flavanone and steroids by cytochrome P450 2C9 variants. Steroids 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-77 28758225-7 2017 All the CYP2C9 variants showed lower flavanone 6-hydroxylation activities than CYP2C9.1 (the wild-type). flavanone 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 28758225-8 2017 CYP2C9.38 showed higher activities in testosterone 6beta-hydroxylation, progesterone 6beta-/16alpha-hydroxylation, estrone 11alpha-hydroxylation and estradiol 6alpha-hydroxylation than CYP2C9.1. Testosterone 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28758225-8 2017 CYP2C9.38 showed higher activities in testosterone 6beta-hydroxylation, progesterone 6beta-/16alpha-hydroxylation, estrone 11alpha-hydroxylation and estradiol 6alpha-hydroxylation than CYP2C9.1. Progesterone 72-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28758225-8 2017 CYP2C9.38 showed higher activities in testosterone 6beta-hydroxylation, progesterone 6beta-/16alpha-hydroxylation, estrone 11alpha-hydroxylation and estradiol 6alpha-hydroxylation than CYP2C9.1. Estrone 115-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28758225-8 2017 CYP2C9.38 showed higher activities in testosterone 6beta-hydroxylation, progesterone 6beta-/16alpha-hydroxylation, estrone 11alpha-hydroxylation and estradiol 6alpha-hydroxylation than CYP2C9.1. Estradiol 149-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28758225-11 2017 These results indicate that the substrate specificity of CYP2C9.9 and CYP2C9.10 was not changed, but CYP2C9.8, CYP2C9.12 and CYP2C9.40 showed different substrate specificity toward steroids compared with CYP2C9.1; and especially CYP2C9.38 displayed diverse substrate specificities towards 7-ethoxycoumarin and steroids. Steroids 181-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 28887089-6 2017 In addition, the inhibitory activities of steviol and stevioside towards the major cytochrome P450 enzymes CYP3A4, CYP2C9, CYP2D6, CYP1A2 and CYP2B6 were evaluated in vitro. steviol 42-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 28887089-6 2017 In addition, the inhibitory activities of steviol and stevioside towards the major cytochrome P450 enzymes CYP3A4, CYP2C9, CYP2D6, CYP1A2 and CYP2B6 were evaluated in vitro. stevioside 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 28887089-8 2017 A weak inhibition of CYP3A4 and CYP2C9 with steviol was also found. steviol 44-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 28893623-2 2017 Abiraterone is known to inhibit several drug metabolizing cytochrome P450 enzymes including CYP1A2, CYP2D6, CYP2C8, CYP2C9, CYP2C19, CYP3A4 and CYP3A5, but its effects on steroid metabolizing P450 enzymes are not clear. abiraterone 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 28834238-0 2017 Long-term outcomes of elderly patients with CYP2C9 and VKORC1 variants treated with vitamin K antagonists. Vitamin K 84-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 28933798-1 2017 Chronic kidney disease (CKD) complicates warfarin anticoagulation partially through its effect on CYP2C9 activity. Warfarin 41-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 28933798-12 2017 CKD increased the effect of CYP2C9 genetic variation on (S)-warfarin and (R,S)-warfarin metabolism. Warfarin 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 28933798-12 2017 CKD increased the effect of CYP2C9 genetic variation on (S)-warfarin and (R,S)-warfarin metabolism. (r,s)-warfarin 73-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 29023392-9 2017 All curcuminoids exhibited multiple pharmacological effects in vitro, including potent antioxidant activity as well as inhibition of CYP2C9, CYP3A4 and lipoxygenase activity without affecting the release of TNF-alpha. curcuminoids 4-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 28609430-0 2017 Polymorphisms in CYP2C9 are associated with response to indomethacin among neonates with patent ductus arteriosus. Indomethacin 56-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 28609430-4 2017 This study investigated the association between single-nucleotide polymorphisms (SNPs) in CYP2C9 and the closure of PDA in response to indomethacin.MethodsSix SNPs in CYP2C9 were analyzed for association with indomethacin response. single-nucleotide 48-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 28609430-4 2017 This study investigated the association between single-nucleotide polymorphisms (SNPs) in CYP2C9 and the closure of PDA in response to indomethacin.MethodsSix SNPs in CYP2C9 were analyzed for association with indomethacin response. Indomethacin 135-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 28609430-4 2017 This study investigated the association between single-nucleotide polymorphisms (SNPs) in CYP2C9 and the closure of PDA in response to indomethacin.MethodsSix SNPs in CYP2C9 were analyzed for association with indomethacin response. methodssix 148-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 28609430-4 2017 This study investigated the association between single-nucleotide polymorphisms (SNPs) in CYP2C9 and the closure of PDA in response to indomethacin.MethodsSix SNPs in CYP2C9 were analyzed for association with indomethacin response. methodssix 148-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 28609430-4 2017 This study investigated the association between single-nucleotide polymorphisms (SNPs) in CYP2C9 and the closure of PDA in response to indomethacin.MethodsSix SNPs in CYP2C9 were analyzed for association with indomethacin response. Indomethacin 209-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 28609430-7 2017 Among indomethacin responders, the G allele of rs2153628 and the T allele of rs1799853 were overtransmitted from the parents to their child (OR: 2.667, 95% CI: 1.374, 5.177 and OR: 2.375, 95% CI: 1.040, 5.425, respectively), consistent with the case-control analysis.ConclusionWe identified an association between two SNPs in CYP2C9, rs2153628 and rs1799853, and indomethacin response for the treatment of PDA. Indomethacin 6-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 326-332 28877533-7 2017 CONCLUSION: Our results further support a minor contribution of CYP2C9 genetic variability toward steady-state endoxifen concentrations. 4-hydroxy-N-desmethyltamoxifen 111-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 28972767-3 2017 Here we report the crystal structures of human CYP2C9 and its polymorphic variants, *3 (I359L) and *30 (A477T), with an antihypertensive drug losartan. Losartan 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 28723731-8 2017 An increase in mean methadone C/D ratios was also seen for homozygous carriers of CYP3A5*3 and heterozygous carriers of CYP2C9*2 or *3 and CYP2C19*2 or *3. Methadone 20-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 28374982-0 2017 Angiotensin II Receptor Blockers Inhibit the Generation of Epoxyeicosatrienoic Acid from Arachidonic Acid in Recombinant CYP2C9, CYP2J2 and Human Liver Microsomes. epoxyeicosatrienoic acid 59-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 28374982-0 2017 Angiotensin II Receptor Blockers Inhibit the Generation of Epoxyeicosatrienoic Acid from Arachidonic Acid in Recombinant CYP2C9, CYP2J2 and Human Liver Microsomes. Arachidonic Acid 89-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-127 28374982-1 2017 Cytochrome P450 (CYP) 2C9, CYP2C8 and CYP2J2 enzymes, which metabolize arachidonic acid (AA) to epoxyeicosatrienoic acids, have cardioprotective effects including anti-inflammation and vasodilation. Arachidonic Acid 71-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 28374982-1 2017 Cytochrome P450 (CYP) 2C9, CYP2C8 and CYP2J2 enzymes, which metabolize arachidonic acid (AA) to epoxyeicosatrienoic acids, have cardioprotective effects including anti-inflammation and vasodilation. epoxyeicosatrienoic acids 96-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 27313202-0 2017 Frequency of Common CYP2C9 Polymorphisms and Their Impact on Warfarin Dose Requirement in Pakistani Population. Warfarin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 27313202-1 2017 Polymorphisms in cytochrome P450 (CYP) 2C9 (CYP2C9) gene result in interindividual variability in warfarin dose requirement. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-42 27313202-1 2017 Polymorphisms in cytochrome P450 (CYP) 2C9 (CYP2C9) gene result in interindividual variability in warfarin dose requirement. Warfarin 98-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 27313202-3 2017 This study was carried out in Pakistani population to evaluate the contribution of common CYP2C9 polymorphisms to warfarin therapy. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 27335128-0 2017 Influence of VKORC1 and CYP2C9 Polymorphisms on Daily Acenocoumarol Dose Requirement in South Indian Patients With Mechanical Heart Valves. Acenocoumarol 54-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 27335128-3 2017 Our aim was to investigate the influence of polymorphisms of VKORC1 and CYP2C9 genes on the mean daily dose requirement of acenocoumarol. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27335128-9 2017 Patients with a wild type of both VKORC1 (-1639GG and 1173CC) and CYP2C9 gene variants required higher acenocoumarol dose compared to those with mutant genotype ( P = 0.023 and P = 0.008 respectively). Acenocoumarol 103-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 27335128-11 2017 CONCLUSION: Presence of a mutant allele of VKORC1 (-1639A & 1173T) and CYP2C9 genes increased the odds of requiring a lower mean dosage of acenocoumarol. Acenocoumarol 143-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 28284562-0 2017 The VKORC1 and CYP2C9 genotypes significantly effect Vitamin K antagonist dosing only in patients over the age of 20years. Vitamin K 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 28934153-9 2017 Selaginellin and selaginellin M also showed medium inhibitory potential against CYP2C9, CYP2J2, UGT1A1, and UGT1A3 (1 muM < IC50 < 5 muM). selaginellin S 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 28934153-9 2017 Selaginellin and selaginellin M also showed medium inhibitory potential against CYP2C9, CYP2J2, UGT1A1, and UGT1A3 (1 muM < IC50 < 5 muM). selaginellin M 17-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 28198005-2 2017 Evidence from the published literature is presented for CYP2C9, VKORC1, CYP4F2, and rs12777823 genotype-guided warfarin dosing to achieve a target international normalized ratio of 2-3 when clinical genotype results are available. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 28748348-9 2017 While CYP2C9*2 and *3 variants significantly reduced phenytoin metabolism, the impacts of CYP2C19*2 and *3 variants were unclear. Phenytoin 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 28927781-0 2017 Function of 38 variants CYP2C9 polymorphism on ketamine metabolism in vitro. Ketamine 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 28435143-5 2017 In this study, we estimated the coefficient of variation (CV) for the intrinsic hepatic clearance of tolbutamide by CYP2C9 for each CYP2C9 genotype using previously reported area under the blood concentration curve (AUC) and oral clearance (CLoral) values in a Monte Carlo simulation with a dispersion model. Tolbutamide 101-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 28435143-5 2017 In this study, we estimated the coefficient of variation (CV) for the intrinsic hepatic clearance of tolbutamide by CYP2C9 for each CYP2C9 genotype using previously reported area under the blood concentration curve (AUC) and oral clearance (CLoral) values in a Monte Carlo simulation with a dispersion model. Tolbutamide 101-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 28435143-6 2017 The CVs for tolbutamide CLint,h,CYP2C9 were estimated to be 18.1%, 23.9%, 25.4%, 22.3%, 13.0%, and 19.8% for CYP2C9*1/*1, *1/*2, *1/*3, *2/*2, *2/*3, and *3/*3, respectively. Tolbutamide 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 28639119-1 2017 PURPOSE: In vitro data showed that selexipag and its active metabolite (ACT-333679) have an inductive effect on CYP3A4, CYP2B6, and CYP2C9 at concentrations approximately 100-fold higher than the maximum plasma concentration (C max) measured under steady-state conditions. selexipag 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 28872889-13 2017 The genotype distributions of the CYP2C9*3, CYP2D6*10, and CYP3A5*3 genetic polymorphisms were associated with the warfarin maintenance dose. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 28872889-15 2017 Moreover, CYP2C9*3, CYP2D6*10, and CYP3A5*3 are associated with the warfarin maintenance dose. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 28927781-5 2017 RESULT: Catalytic activity of thirty-eight CYP2C9 alleles on ketamine metabolism to norketamine was surveyed. Ketamine 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 28927781-5 2017 RESULT: Catalytic activity of thirty-eight CYP2C9 alleles on ketamine metabolism to norketamine was surveyed. norketamine 84-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 28820457-0 2017 Role of CYP2C9, CYP2C19 and EPHX Polymorphism in the Pharmacokinetic of Phenytoin: A Study on Uruguayan Caucasian Subjects. Phenytoin 72-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 29156869-4 2017 A warfarin dosing algorithm was developed based on age, height, CYP2C9, VKORC1, and GGCX genotype. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 29181084-2 2017 Patients & methods: Test results for three common cardiovascular drug-gene tests (warfarin-CYP2C9-VKORC1, clopidogrel-CYP2C19 and simvastatin-SLCO1B1) of 208 patients in the Ohio State University-Coriell Personalized Medicine Collaborative were examined to determine the incidence of potentially actionable test results. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 28841884-18 2017 UGT1A6/CYP2C9 polymorphisms have influence on VPA metabolism. Valproic Acid 46-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 7-13 28820457-1 2017 Phenytoin (PHT) oxidative route leads to its main metabolite p-hydroxyphenytoin (p-HPPH), by means of CYP2C9 and CYP2C19. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 28820457-1 2017 Phenytoin (PHT) oxidative route leads to its main metabolite p-hydroxyphenytoin (p-HPPH), by means of CYP2C9 and CYP2C19. Phenytoin 11-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 28820457-1 2017 Phenytoin (PHT) oxidative route leads to its main metabolite p-hydroxyphenytoin (p-HPPH), by means of CYP2C9 and CYP2C19. hydroxyphenytoin 61-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 28820457-1 2017 Phenytoin (PHT) oxidative route leads to its main metabolite p-hydroxyphenytoin (p-HPPH), by means of CYP2C9 and CYP2C19. p-hpph 81-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 28429387-2 2017 Polymorphisms in the VKORC1, CYP2C9 and CYP4F2 genes have been verified to correlate with warfarin stable dosage (WSD). Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 28550460-6 2017 Similarly, the patients with CYP2C9*3 variant allele required significantly lower warfarin SWDs than those with homozygous wild-type (p = 0.006). Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 29156869-7 2017 A warfarin dosing algorithm showed that age, height, CYP2C9, VKORC1, and GGCX genotype were the best variables for estimating warfarin dose (R2 = 41.2%). Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 28273397-0 2017 Associations of CYP2C9 and CYP2A6 Polymorphisms with the Concentrations of Valproate and its Hepatotoxin Metabolites and Valproate-Induced Hepatotoxicity. Valproic Acid 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 28273397-0 2017 Associations of CYP2C9 and CYP2A6 Polymorphisms with the Concentrations of Valproate and its Hepatotoxin Metabolites and Valproate-Induced Hepatotoxicity. Valproic Acid 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 27853934-8 2017 RESULTS: N-oxides of clozapine (CYP2B6/2C19) and voriconazole (CYP2C9/3A4) showed CYP inhibition >=50%. n-oxides 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 27853934-8 2017 RESULTS: N-oxides of clozapine (CYP2B6/2C19) and voriconazole (CYP2C9/3A4) showed CYP inhibition >=50%. Clozapine 21-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 27853934-8 2017 RESULTS: N-oxides of clozapine (CYP2B6/2C19) and voriconazole (CYP2C9/3A4) showed CYP inhibition >=50%. Voriconazole 49-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 28608988-0 2017 Dosing algorithms for vitamin K antagonists across VKORC1 and CYP2C9 genotypes: comment. Vitamin K 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 28604475-10 2017 RESULTS: First, the concomitant use of carbapenems (beta" = -0.422) and UGT1A6 (552 AA AC) (beta" = -0.249) had a significant negative correlation with the weight-adjusted VPA serum concentration (C:W ratio), whereas CYP2C9 (1075 AA AC) (beta" = 0.186) and gender (female compared with male) (beta" = 0.322) showed a positive correlation with VPA serum C:W ratio. Carbapenems 39-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 28638342-7 2017 The CYP2C9 variant rs2860905, which was found in all the major haplotypes occurring in the Puerto Rican population, showed stronger association with warfarin sensitivity (<4 mg/day) than common variants CYP2C9*2 and CYP2C9*3. Warfarin 149-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 28805981-3 2017 Another aim was to examine the in vitro influence of ethanol on the CYP2C9 activity. Ethanol 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 28805981-4 2017 Human CYP2C9 (wild type) isolated from a baculovirus-infected cell system was incubated with 0.8 mumol/L losartan for 20 min. Losartan 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 28805981-7 2017 Formation of the CYP2C9-produced losartan metabolite EXP-3174 was determined by validated LC-MS/MS methodology. Losartan 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 28805981-15 2017 The presence of ethanol inhibited CYP2C9 activity in a concentration-dependent manner. Ethanol 16-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 28744217-14 2017 Article summary: (1) Children carrying functional alleles in GSTM1 and CYP2C9 are at high risk for developing hemorrhagic cystitis following treatment with busulfan and cyclophosphamide based conditioning regimen. Busulfan 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 28744217-14 2017 Article summary: (1) Children carrying functional alleles in GSTM1 and CYP2C9 are at high risk for developing hemorrhagic cystitis following treatment with busulfan and cyclophosphamide based conditioning regimen. Cyclophosphamide 169-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 28685643-0 2017 Clinical effect of CYP2C9*5/*6 genotype on a patient"s warfarin dose requirement. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 28685643-1 2017 We describe a 38-year-old African-American female treated with warfarin for acute bilateral pulmonary emboli who is a carrier of two rare CYP2C9 variant alleles, *5 and *6, along with VKORC1 -1639GG and CYP4F2 433Val/Val genotypes. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 28685643-3 2017 This case sheds further light on the cumulative clinical impact of the CYP2C9 variant alleles, *5 and *6, on warfarin dose requirements and practical considerations for warfarin genotyping in a racially and ethnically diverse population. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 28685643-3 2017 This case sheds further light on the cumulative clinical impact of the CYP2C9 variant alleles, *5 and *6, on warfarin dose requirements and practical considerations for warfarin genotyping in a racially and ethnically diverse population. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 28740425-0 2017 Efficacy of piroxicam for postoperative pain after lower third molar surgery associated with CYP2C8*3 and CYP2C9. Piroxicam 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 28740425-2 2017 The aim of this study was to evaluate the possible association of polymorphisms in the CYP2C8*3 and CYP2C9 genes with the clinical efficacy of oral piroxicam (20 mg daily for 4 days) after lower third molar surgeries with regard to postoperative pain, swelling, trismus, adverse reactions, need for rescue medication and the volunteer"s overall satisfaction. Piroxicam 148-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 28283499-6 2017 CYP2B6 and to an even lesser extent CYP2C9 were also observed to catalyze NGMN metabolism. ngmn 74-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 28370504-0 2017 CYP1A1m1 and CYP2C9*2 and *3 polymorphism and risk to develop ARV-associated hepatotoxicity and its severity. omega-N-Allylarginine 62-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 28370504-8 2017 CYP2C9*31075AC genotype among alcohol users was likely to be associated with development of ARV-associated hepatotoxicity (OR = 1.67, p = 0.38). Alcohols 30-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28370504-8 2017 CYP2C9*31075AC genotype among alcohol users was likely to be associated with development of ARV-associated hepatotoxicity (OR = 1.67, p = 0.38). omega-N-Allylarginine 92-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28370504-11 2017 CYP1A1m1 3801TC, 3801CC, and CYP2C9*3 1075AC genotypes among combined alcohol + nevirapine users increased the risk of development of ARV-associated hepatotoxicity (OR = 1.41, p = 0.53; OR = 1.49, p = 0.83; OR = 1.78, p = 0.35). Alcohols 70-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 28370504-11 2017 CYP1A1m1 3801TC, 3801CC, and CYP2C9*3 1075AC genotypes among combined alcohol + nevirapine users increased the risk of development of ARV-associated hepatotoxicity (OR = 1.41, p = 0.53; OR = 1.49, p = 0.83; OR = 1.78, p = 0.35). Nevirapine 80-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 28370504-11 2017 CYP1A1m1 3801TC, 3801CC, and CYP2C9*3 1075AC genotypes among combined alcohol + nevirapine users increased the risk of development of ARV-associated hepatotoxicity (OR = 1.41, p = 0.53; OR = 1.49, p = 0.83; OR = 1.78, p = 0.35). omega-N-Allylarginine 134-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 28370504-13 2017 CYP2C9*31075AC genotype with combined alcohol and nevirapine usage indicated a risk for development of ARV-associated hepatotoxicity. Alcohols 38-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28370504-13 2017 CYP2C9*31075AC genotype with combined alcohol and nevirapine usage indicated a risk for development of ARV-associated hepatotoxicity. Nevirapine 50-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28484907-4 2017 MAM-2201 potently inhibited CYP2C9-catalyzed diclofenac 4"-hydroxylation, CYP3A4-catalyzed midazolam 1"-hydroxylation, and UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-glucuronidation, with K i values of 5.6, 5.4 and 5.0 microM, respectively. Diclofenac 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 28114735-2 2017 Carvedilol is mainly metabolized by glucuronidation, by CYP2D6 to hydroxyphenylcarvedilol (OHC), and by CYP2C9 to O-desmethylcarvedilol (DMC). Carvedilol 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28262345-1 2017 OBJECTIVES: The aim of this study was to evaluate the associations between polymorphisms of VKORC1, CYP2C9, CYP4F2, NR3C1 and VDR genes and stable warfarin doses in Korean patients with mechanical heart valves. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 28262345-9 2017 CONCLUSIONS: This study showed that stable warfarin dose is associated with VDR SNPs along with VKORC1, CYP2C9, and CYP4F2 SNPs. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28114735-2 2017 Carvedilol is mainly metabolized by glucuronidation, by CYP2D6 to hydroxyphenylcarvedilol (OHC), and by CYP2C9 to O-desmethylcarvedilol (DMC). desmethylcarvedilol 114-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28114735-2 2017 Carvedilol is mainly metabolized by glucuronidation, by CYP2D6 to hydroxyphenylcarvedilol (OHC), and by CYP2C9 to O-desmethylcarvedilol (DMC). desmethylcarvedilol 137-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 28626479-1 2017 The genetic factors are determinants in required dosage changes of warfarin among which are polymorphisms of CYP2C9 and VKORC1 genes. Warfarin 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 28412319-2 2017 We hypothesized that genetic factors related to warfarin metabolism (CYP2C9) and activity (VKORC1) would show stronger associations than modifiable factors with the quality of anticoagulation control and risks for thromboembolism and hemorrhage. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 28284082-1 2017 Cytochrome P450 (CYP) 2C9 and CYP2C19 genetic mutant could influence the plasma concentration of glipizide in human subjects, which refers to glipizide safety and adverse effects in clinic practice. Glipizide 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 28284082-1 2017 Cytochrome P450 (CYP) 2C9 and CYP2C19 genetic mutant could influence the plasma concentration of glipizide in human subjects, which refers to glipizide safety and adverse effects in clinic practice. Glipizide 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 27665573-0 2017 Evaluation of Concomitant Antiretrovirals and CYP2C9/CYP2C19 Polymorphisms on the Pharmacokinetics of Etravirine. etravirine 102-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 27665573-2 2017 Etravirine is primarily metabolized by cytochrome P450 (CYP) 2C9, CYP2C19, and CYP3A. etravirine 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-64 28032893-2 2017 Utilizing data from a diverse population of 257 patients who received VKORC1 and CYP2C9 genotype-guided warfarin dosing, we aimed to examine genotype-associated differences in anticoagulation endpoints and derive a novel pharmacogenetic nomogram to more optimally dose warfarin. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 27665573-3 2017 This analysis determined the impact of concomitant antiretrovirals and CYP2C9/CYP2C19 phenotype on the pharmacokinetics of etravirine. etravirine 123-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 27665573-10 2017 CL/F increased non-linearly with body weight and creatinine clearance (CLCR), and also varied based on CYP2C9/CYP2C19 phenotype. Fluorine 3-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 27665573-11 2017 CONCLUSIONS: In this analysis, body weight, CLCR, and CYP2C9/CYP2C19 phenotype were found to describe some of the variability in CL/F. Fluorine 132-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 28315807-0 2017 Association of CYP2C9, CYP2A6, ACSM2A, and CPT1A gene polymorphisms with adverse effects of valproic acid in Chinese patients with epilepsy. Valproic Acid 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 28315807-5 2017 RESULTS: CYP2C9 mutations had a significant impact on 4-ene-VPA concentration, in patients with wild-type CYP2C9 (CYP2C9*1), which has a greater capacity for VPA metabolism than the mutant type (CYP2C9*3), liver dysfunction was substantially higher. 4-ene 54-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 28315807-5 2017 RESULTS: CYP2C9 mutations had a significant impact on 4-ene-VPA concentration, in patients with wild-type CYP2C9 (CYP2C9*1), which has a greater capacity for VPA metabolism than the mutant type (CYP2C9*3), liver dysfunction was substantially higher. 4-ene 54-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 28173639-5 2017 In primary human hepatocytes, CYP2C9 and CYP3A4 gene expression and activities decreased upon treatment with 20 muM bosentan+200 muM rifampin. Bosentan 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 28173639-5 2017 In primary human hepatocytes, CYP2C9 and CYP3A4 gene expression and activities decreased upon treatment with 20 muM bosentan+200 muM rifampin. Rifampin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 28315807-1 2017 OBJECTIVE: To explore the influence of CYP2C9, CYP2A6, ACSM2A, CPT1A gene polymorphisms on valproic acid (VPA) and its role in metabolism-related liver dysfunction in order to guide the clinical safety and rational use of VPA. Valproic Acid 91-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 28315807-5 2017 RESULTS: CYP2C9 mutations had a significant impact on 4-ene-VPA concentration, in patients with wild-type CYP2C9 (CYP2C9*1), which has a greater capacity for VPA metabolism than the mutant type (CYP2C9*3), liver dysfunction was substantially higher. 4-ene 54-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 28315807-5 2017 RESULTS: CYP2C9 mutations had a significant impact on 4-ene-VPA concentration, in patients with wild-type CYP2C9 (CYP2C9*1), which has a greater capacity for VPA metabolism than the mutant type (CYP2C9*3), liver dysfunction was substantially higher. 4-ene 54-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 28315807-1 2017 OBJECTIVE: To explore the influence of CYP2C9, CYP2A6, ACSM2A, CPT1A gene polymorphisms on valproic acid (VPA) and its role in metabolism-related liver dysfunction in order to guide the clinical safety and rational use of VPA. Valproic Acid 106-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 28157069-0 2017 Delayed de-induction of CYP2C9 compared to CYP3A after discontinuation of rifampicin: Report of two cases : . Rifampin 74-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 28157069-3 2017 To clarify the time courses of deinduction of CYP2C9 and -CYP3A activities after rifampicin withdrawal, we monitored these enzyme activities in 2 patients over time after discontinuing rifampicin. Rifampin 81-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 28157069-3 2017 To clarify the time courses of deinduction of CYP2C9 and -CYP3A activities after rifampicin withdrawal, we monitored these enzyme activities in 2 patients over time after discontinuing rifampicin. Rifampin 185-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 28157069-7 2017 We monitored CYP2C9 and CYP3A activities over their clinical courses by measuring the metabolic clearance of S-warfarin to S-7-hydroxywarfarin and that of cortisol to 6beta-hydroxycortisol, respectively. Warfarin 109-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 28157069-7 2017 We monitored CYP2C9 and CYP3A activities over their clinical courses by measuring the metabolic clearance of S-warfarin to S-7-hydroxywarfarin and that of cortisol to 6beta-hydroxycortisol, respectively. Hydrocortisone 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 28157069-7 2017 We monitored CYP2C9 and CYP3A activities over their clinical courses by measuring the metabolic clearance of S-warfarin to S-7-hydroxywarfarin and that of cortisol to 6beta-hydroxycortisol, respectively. 6 beta-hydroxycortisol 167-188 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 27826892-0 2017 The effect of UGT1A9, CYP2B6 and CYP2C9 genes polymorphism on individual differences in propofol pharmacokinetics among Polish patients undergoing general anaesthesia. Propofol 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 28468305-5 2017 Dimethyllirioresinol weakly inhibited CYP2C19 and CYP2C8 with IC50 values of 55.1 and 85.0 muM, respectively, without inhibition of CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, and CYP3A4 activities at 100 muM. dimethyllirioresinol 0-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 27826892-4 2017 The aim of our study was to verify the impact of genetic changes c.516G>T in the CYP2B6, c.98T>C in the UGT1A9 and c.1075A>C in the CYP2C9 genes on the individual propofol pharmacokinetic profile in the Polish patients undergoing general anaesthesia. Propofol 172-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 28073118-9 2017 The main human cytochrome P450 isoforms involved in Artepillin C metabolism had been identified, and the results suggest a majority contribution of CYP2E1 and CYP2C9 in the formation of the two metabolites. artepillin C 52-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 159-165 28073119-4 2017 The apparent Ki value for CYP2C9 was 50.60 microM and those for CYP3A4/5 were 48.71 microM and 31.25 microM using two different probe substrates, nifedipine and midazolam, respectively. Nifedipine 146-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 28073119-4 2017 The apparent Ki value for CYP2C9 was 50.60 microM and those for CYP3A4/5 were 48.71 microM and 31.25 microM using two different probe substrates, nifedipine and midazolam, respectively. Midazolam 161-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 28520385-3 2012 Prasugrel is metabolized to its active metabolite primarily by CYP3A5 and CYP2B6, and to a lesser extent by CYP2C9 and CYP2C19. Prasugrel Hydrochloride 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 28484392-0 2017 "Late" Withdrawal Syndrome after Carbamazepine In Utero Exposure in a CYP2C9 Slow Metabolizer Newborn. Carbamazepine 33-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 28484392-5 2017 The infant genotype analysis showed two low activity CYP2C9 allelic variants (*2/*3 heterozygote) predicting a CYP2C9 slow metabolizer phenotype which could explain reduced carbamazepine elimination and a late and long-lasting withdrawal symptoms observed 3 days after birth. Carbamazepine 173-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 28484392-5 2017 The infant genotype analysis showed two low activity CYP2C9 allelic variants (*2/*3 heterozygote) predicting a CYP2C9 slow metabolizer phenotype which could explain reduced carbamazepine elimination and a late and long-lasting withdrawal symptoms observed 3 days after birth. Carbamazepine 173-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 28274629-4 2017 6-Paradol showed concentration-dependent inhibitory effects on CYP1A2, CYP2B6, CYP2C8, CYP2C9, and CYP2C19 isozymes, with IC50 values ranging from 3.8 to 21.4microM in recombinant CYP isozymes. 6-paradol 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27763679-9 2017 CYP2C9 *1/*3 genotype had reduced clearance for S-warfarin, but increased clearance for R-warfarin. Sulfur 48-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27763679-9 2017 CYP2C9 *1/*3 genotype had reduced clearance for S-warfarin, but increased clearance for R-warfarin. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27763679-9 2017 CYP2C9 *1/*3 genotype had reduced clearance for S-warfarin, but increased clearance for R-warfarin. Warfarin 88-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 28099407-2 2017 We investigated whether the variant genotypes of cytochrome P450 2C8 (CYP2C8), CYP2C9, CYP3A5 and transporter ABCB1 influence the pharmacokinetic phenotype of the substrate pioglitazone in Chinese individuals. Pioglitazone 173-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 28135601-8 2017 On the other hand, tested flavonoids showed no or weaker inhibition of CYP2C9 compared to warfarin, making it very unlikely that quercetin or its metabolites can significantly inhibit the CYP2C9-mediated inactivation of warfarin. Warfarin 220-228 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 28135054-7 2017 Pharmacogenomics-guided warfarin dosing algorithms that include the well-established VKORC1 and CYP2C9 polymorphisms explain only a small proportion of inter-individual variability in African Americans (AAs). Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 28135054-11 2017 Inclusion of rs4889606 genotypes, along with CYP2C9 alleles, rs9923231 genotypes and clinical variables, explained 31% of the inter-patient variability in warfarin dose requirement. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 28135601-0 2017 Interaction of quercetin and its metabolites with warfarin: Displacement of warfarin from serum albumin and inhibition of CYP2C9 enzyme. Quercetin 15-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 28135601-0 2017 Interaction of quercetin and its metabolites with warfarin: Displacement of warfarin from serum albumin and inhibition of CYP2C9 enzyme. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 28135601-5 2017 Furthermore, the inhibitory effects of these flavonoids on CYP2C9 enzyme were tested because the metabolic elimination of warfarin is catalysed principally by this enzyme. Flavonoids 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 28179134-3 2017 The results demonstrate that most of the unsaturated fatty acids showed marked inhibition towards CYP2C8 mediated amodiaquine N-deethylation followed by inhibition of CYP2C9 and CYP2B6 mediated activities. Fatty Acids, Unsaturated 41-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 30455562-0 2017 Warfarin maintenance dose associated with genetic polymorphisms of CYP2C9. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 30455562-1 2017 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) gene polymorphisms alters the required warfarin dose in patients, due to pharmacogenetic events. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-31 30455562-1 2017 BACKGROUND: Cytochrome P450 2C9 (CYP2C9) gene polymorphisms alters the required warfarin dose in patients, due to pharmacogenetic events. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 30455562-9 2017 CONCLUSIONS: Due to the relatively high frequency of CYP2C9 polymorphisms in the study population, the clinicians should become aware of these results to reduce the risk of hemorrhage when prescribing warfarin. Warfarin 201-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 28129313-2 2017 Some SSRIs are potent CYP2C9 inhibitors that may increase warfarin plasma concentrations and the risk of bleeding. Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 28129313-3 2017 We aimed to examine the effect of the putative CYP2C9-mediated warfarin-SSRI interaction on clinical outcomes. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 28129313-10 2017 CONCLUSIONS: Patients concomitantly treated with warfarin and SSRIs that are potent CYP2C9 inhibitors had comparable rates of bleeding events, ischemic or thromboembolic events, and mortality as did patients cotreated with warfarin and other SSRIs, although small but potentially meaningful effects on bleeding cannot be completely excluded. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 28099407-7 2017 Unexpectedly, the pioglitazone AUC0-48 in CYP2C9*3 carriers was lower (50.8%), whereas the AUC0-48 ratios of metabolites M-III/pioglitazone and M-IV/pioglitazone increased to 134.3 and 155.8%, respectively, compared with the wild-type CYP2C9*1/*1 homozygous. Pioglitazone 18-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 27312150-5 2017 The most potent inhibitors were genistein and daidzein inhibiting noncompetitively the CYP2C9 with Ki of 35.95 +- 6.96 and 60.56 +- 3.53 mumol/l and CYP3A4 (inhibited by genistein with Ki of 23.25 +- 5.85 mumol/l also by a noncompetitive mechanism). Genistein 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27312150-5 2017 The most potent inhibitors were genistein and daidzein inhibiting noncompetitively the CYP2C9 with Ki of 35.95 +- 6.96 and 60.56 +- 3.53 mumol/l and CYP3A4 (inhibited by genistein with Ki of 23.25 +- 5.85 mumol/l also by a noncompetitive mechanism). daidzein 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27312150-5 2017 The most potent inhibitors were genistein and daidzein inhibiting noncompetitively the CYP2C9 with Ki of 35.95 +- 6.96 and 60.56 +- 3.53 mumol/l and CYP3A4 (inhibited by genistein with Ki of 23.25 +- 5.85 mumol/l also by a noncompetitive mechanism). Genistein 170-179 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27312150-8 2017 Genistein and daidzein inhibit noncompetitively CYP3A4 and CYP2C9. Genistein 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 27312150-8 2017 Genistein and daidzein inhibit noncompetitively CYP3A4 and CYP2C9. daidzein 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 27864660-4 2017 A single 200-mg oral dose of celecoxib was given to 52 Korean subjects with different CYP2C9 genotypes: CYP2C9EM (n = 26; CYP2C9*1/*1), CYP2C9IM (n = 24; CYP2C9*1/*3 and *1/*13), and CYP2C9PM (n = 2; CYP2C9*3/*3). Celecoxib 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 28287454-6 2017 These in vitro results indicate that AM-2201 needs to be examined for potential pharmacokinetic drug interactions in vivo due to its potent inhibition of CYP2C8, CYP2C9, CYP3A4, UGT1A3, and UGT2B7 enzyme activities. 1-(5-fluoropentyl)-3-(1-naphthoyl)indole 37-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 27864660-0 2017 Effects of CYP2C9 genetic polymorphisms on the pharmacokinetics of celecoxib and its carboxylic acid metabolite. Celecoxib 67-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 27864660-2 2017 The predominant hepatic metabolism of celecoxib to celecoxib carboxylic acid (CCA) is mediated mainly by CYP2C9. Celecoxib 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 28408837-0 2017 Polymorphisms of VKORC1 and CYP2C9 are associated with warfarin sensitivity in Chinese population. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 28408837-3 2017 The present study was designed to clarify the real biological significance of the polymorphisms of VKORC1 and cytochrome P450 2C9 (CYP2C9) in warfarin metabolism. Warfarin 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-129 28408837-3 2017 The present study was designed to clarify the real biological significance of the polymorphisms of VKORC1 and cytochrome P450 2C9 (CYP2C9) in warfarin metabolism. Warfarin 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 28408837-8 2017 VKORC1 1173CT, 1542GC, 2255CT, 3730CT carriers and CYP2C9 exon 4 -65GC carriers had significantly higher warfarin daily dosage than others (3.2+-0.6 vs 3.1+-1.1 vs 2.6+-0.8 mg/day). Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 28083852-0 2017 DNA sensors to assess the effect of VKORC1 and CYP2C9 gene polymorphisms on warfarin dose requirement in Chinese patients with atrial fibrillation. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 28083852-1 2017 The optimal dose of warfarin depends on polymorphisms in the VKORC1 (the vitamin K epoxide reductase complex subunit (1) and CYP2C9 (cytochrome P450 2C9) genes. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 28083852-1 2017 The optimal dose of warfarin depends on polymorphisms in the VKORC1 (the vitamin K epoxide reductase complex subunit (1) and CYP2C9 (cytochrome P450 2C9) genes. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-152 27864660-4 2017 A single 200-mg oral dose of celecoxib was given to 52 Korean subjects with different CYP2C9 genotypes: CYP2C9EM (n = 26; CYP2C9*1/*1), CYP2C9IM (n = 24; CYP2C9*1/*3 and *1/*13), and CYP2C9PM (n = 2; CYP2C9*3/*3). Celecoxib 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 27864660-2 2017 The predominant hepatic metabolism of celecoxib to celecoxib carboxylic acid (CCA) is mediated mainly by CYP2C9. SC-62807 51-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 27864660-2 2017 The predominant hepatic metabolism of celecoxib to celecoxib carboxylic acid (CCA) is mediated mainly by CYP2C9. 1-methylcyclohexanecarboxylic acid 78-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 27864660-3 2017 We investigated the effects of the major CYP2C9 genetic variants in Asian populations, CYP2C9*3 and CYP2C9*13, on the pharmacokinetics of celecoxib and its carboxylic acid metabolite in healthy Korean subjects. Celecoxib 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 27864660-3 2017 We investigated the effects of the major CYP2C9 genetic variants in Asian populations, CYP2C9*3 and CYP2C9*13, on the pharmacokinetics of celecoxib and its carboxylic acid metabolite in healthy Korean subjects. Celecoxib 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27864660-3 2017 We investigated the effects of the major CYP2C9 genetic variants in Asian populations, CYP2C9*3 and CYP2C9*13, on the pharmacokinetics of celecoxib and its carboxylic acid metabolite in healthy Korean subjects. Celecoxib 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 27864660-4 2017 A single 200-mg oral dose of celecoxib was given to 52 Korean subjects with different CYP2C9 genotypes: CYP2C9EM (n = 26; CYP2C9*1/*1), CYP2C9IM (n = 24; CYP2C9*1/*3 and *1/*13), and CYP2C9PM (n = 2; CYP2C9*3/*3). Celecoxib 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 27864660-4 2017 A single 200-mg oral dose of celecoxib was given to 52 Korean subjects with different CYP2C9 genotypes: CYP2C9EM (n = 26; CYP2C9*1/*1), CYP2C9IM (n = 24; CYP2C9*1/*3 and *1/*13), and CYP2C9PM (n = 2; CYP2C9*3/*3). Celecoxib 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 27864660-6 2017 The mean area under the plasma concentration-time curve (AUC0- ) of celecoxib was increased 1.63-fold (P < 0.001), and the apparent oral clearance (CL/F) of celecoxib was decreased by 39.6% in the CYP2C9IM genotype group compared with that of CYP2C9EM (P < 0.001). Celecoxib 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 200-206 27864660-7 2017 The overall pharmacokinetic parameters for celecoxib in CYP2C9*1/*13 subjects were similar to those in CYP2C9*1/*3 subjects. Celecoxib 43-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 27864660-8 2017 Two subjects with CYP2C9PM genotype both showed markedly higher AUC0- , prolonged half-life, and lower CL/F for celecoxib than did subjects with CYP2C9EM and IM genotypes. Celecoxib 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 27864660-8 2017 Two subjects with CYP2C9PM genotype both showed markedly higher AUC0- , prolonged half-life, and lower CL/F for celecoxib than did subjects with CYP2C9EM and IM genotypes. Celecoxib 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 27864660-9 2017 CYP2C9*3 and CYP2C9*13 variant alleles significantly affected the plasma concentration of celecoxib. Celecoxib 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27864660-9 2017 CYP2C9*3 and CYP2C9*13 variant alleles significantly affected the plasma concentration of celecoxib. Celecoxib 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 28074333-7 2017 The CYP2C8 (69 +- 20%), CYP2C9 (42 +- 10%), CYP3A4 (52 +- 23%), and CEP2E1 (41 +- 13%) inhibitors all significantly inhibited rosiglitazone metabolism. Rosiglitazone 126-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 27718269-11 2017 High expression of CYP3A4, CYP2C8, CYP2C9, and CYP2D6 may be risk factors for hepatotoxicity caused by AQ-therapy. Amodiaquine 103-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 28074333-8 2017 CONCLUSION: The results suggest that other cytochrome P450 enzymes, including CYP2C9, CYP3A4, and CEP2E1, in addition to CYP28, also play an important role in the metabolism of rosiglitazone. Rosiglitazone 177-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 28063245-0 2017 Dosing algorithms for vitamin K antagonists across VKORC1 and CYP2C9 genotypes. Vitamin K 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 27273787-0 2017 Upregulation of human CYP2C9 expression by Bisphenol A via estrogen receptor alpha (ERalpha) and Med25. bisphenol A 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 27273787-5 2017 We further identify the mechanism of BPA upregulation of CYP2C9 expression. bisphenol A 37-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27273787-8 2017 Our findings suggest that BPA exposure has a potential risk for adverse health effects in human liver metabolism by upregulation of CYP2C9 expression. bisphenol A 26-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 28063245-13 2017 Conclusions EU-PACT genetic-guided dose initiation algorithms for acenocoumarol and phenprocoumon could have predicted the dose overcautiously in the VKORC1 AA-CYP2C9*1*1 subgroup. Acenocoumarol 66-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 27539372-0 2017 CYP2C9 Genotype-Dependent Warfarin Pharmacokinetics: Impact of CYP2C9 Genotype on R- and S-Warfarin and Their Oxidative Metabolites. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27539372-0 2017 CYP2C9 Genotype-Dependent Warfarin Pharmacokinetics: Impact of CYP2C9 Genotype on R- and S-Warfarin and Their Oxidative Metabolites. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 27539372-0 2017 CYP2C9 Genotype-Dependent Warfarin Pharmacokinetics: Impact of CYP2C9 Genotype on R- and S-Warfarin and Their Oxidative Metabolites. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27539372-0 2017 CYP2C9 Genotype-Dependent Warfarin Pharmacokinetics: Impact of CYP2C9 Genotype on R- and S-Warfarin and Their Oxidative Metabolites. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 27539372-1 2017 Multiple factors can impact warfarin therapy, including genetic variations in the drug-metabolizing enzyme cytochrome P450 2C9 (CYP2C9). Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-126 27539372-1 2017 Multiple factors can impact warfarin therapy, including genetic variations in the drug-metabolizing enzyme cytochrome P450 2C9 (CYP2C9). Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 27539372-3 2017 Genetic variations in CYP2C9, the primary enzyme governing the metabolic clearance of the more potent S-enantiomer of the racemic anticoagulant warfarin, may impact warfarin-drug interactions. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 27539372-3 2017 Genetic variations in CYP2C9, the primary enzyme governing the metabolic clearance of the more potent S-enantiomer of the racemic anticoagulant warfarin, may impact warfarin-drug interactions. Warfarin 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 20-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-5 2017 Plasma clearance of S-warfarin, but not R-warfarin, decreased multiexponentially and in a CYP2C9 gene-dependent manner: 56%, 70%, and 75% for CYP2C9*1/*3, CYP2C9*2/*3, and CYP2C9*3/*3 genotypes, respectively, compared with CYP2C9*1/*1, resulting in pronounced differences in the S:R ratio that identified warfarin-sensitive genotypes. Warfarin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 142-148 27539372-6 2017 CYP2C9 was the primary P450 enzyme contributing to S-warfarin metabolism and a minor contributor to R-warfarin metabolism. Sulfur 51-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27539372-6 2017 CYP2C9 was the primary P450 enzyme contributing to S-warfarin metabolism and a minor contributor to R-warfarin metabolism. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27539372-6 2017 CYP2C9 was the primary P450 enzyme contributing to S-warfarin metabolism and a minor contributor to R-warfarin metabolism. Warfarin 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27539372-7 2017 In the presence of a defective CYP2C9 allele, switching of warfarin metabolism to other oxidative pathways and P450 enzymes for the metabolic elimination of S-warfarin was not observed. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 27539372-7 2017 In the presence of a defective CYP2C9 allele, switching of warfarin metabolism to other oxidative pathways and P450 enzymes for the metabolic elimination of S-warfarin was not observed. Warfarin 159-167 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 27539372-9 2017 Understanding the impact of CYP2C9 genetics on warfarin pharmacokinetics lays the foundation for future genotype-dependent warfarin-drug interaction studies. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 27539372-9 2017 Understanding the impact of CYP2C9 genetics on warfarin pharmacokinetics lays the foundation for future genotype-dependent warfarin-drug interaction studies. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 28224948-10 2017 Patients, however, homozygous for CYP2C9 *1,*3, and VKORC1 required less than 5 mg/day of warfarin to maintain the INR within the therapeutic range. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 28649054-0 2017 Effect of VKORC1 and CYP2C9 polymorphisms on warfarin dose requirement in Bangladeshi population. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 28649054-2 2017 Large inter-individuals variability due to age, gender, diet, concurrent drug interactions and variations in CYP2C9 and VKORC1 genes make the management of warfarin therapy challenging and yet no study has been conducted on the Bangladeshi population. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 28649054-3 2017 The aim of the study was to identify the role of VKORC1 and CYP2C9 polymorphisms in Bangladeshi population in dose requirement of warfarin. Warfarin 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 28649054-8 2017 The results of this study indicate that comparatively higher daily maintenance doses of warfarin were required to achieve the target INR for patients carrying both GG genotype of VKORC1rs9923231 and wild type variant of CYP2C9*3 whereas minimum dose were required for patient having AA genotype of VKORC1rs9923231 and *3/*3 variant of CYP2C9. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 28649054-8 2017 The results of this study indicate that comparatively higher daily maintenance doses of warfarin were required to achieve the target INR for patients carrying both GG genotype of VKORC1rs9923231 and wild type variant of CYP2C9*3 whereas minimum dose were required for patient having AA genotype of VKORC1rs9923231 and *3/*3 variant of CYP2C9. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 335-341 27934636-7 2017 Only using the RAF derived from testosterone for CYP3A4 produced the expected CYP2C9 contribution of 72%-87% and 47%-69% for metabolism of losartan and meloxicam, respectively. Meloxicam 152-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27934636-4 2017 The RAF value for CYP3A4 or CYP2C9 derived from a particular P450-selective probe reaction was applied to calculate RAF-scaled CLint for other probe reactions of the same P450 isoform in a crossover manner and compared with the measured HLM CLint When RAF derived from midazolam or nifedipine was used for CYP3A4, the ICR for testosterone 6beta-hydroxylation was 31 and 25, respectively, suggesting significantly diverse interactions of CYP3A4 probes with the testing systems. Midazolam 269-278 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 27661060-1 2017 AIM: To assess the effect of Cytochrome P450 2C9 (CYP2C9) gene polymorphism on pediatric warfarin maintenance dosage requirement. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-48 27661060-1 2017 AIM: To assess the effect of Cytochrome P450 2C9 (CYP2C9) gene polymorphism on pediatric warfarin maintenance dosage requirement. Warfarin 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 27661060-5 2017 Review Manager version 5.2.3 software was used to analyze the relationship between CYP2C9 polymorphisms and warfarin maintenance doses in pediatric patients. Warfarin 108-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 27661060-7 2017 RESULTS: Maintenance warfarin doses in patients with CYP2C9 *1/*2 genotype, CYP2C9 *1/*3 genotype, and CYP2C9 variant carriers which contain at least one variant allele (*2 or *3) were from 15% to 41% lower than doses in patients with the wild-type allele (CYP2C9 *1/*1): All differences were significant with P-values <.05. Warfarin 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 27661060-9 2017 CONCLUSIONS: We found that CYP2C9 gene polymorphism (referring to the presence of *1/*2, *1/*3, and variant genotypes in the population in addition to the wild type) was significantly associated with decreased warfarin maintenance dose requirements. Warfarin 210-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 28131653-7 2017 We also found that the 6-hydroxylation of 1"-hydroxy BBR is mainly catalyzed by CYP2C9 and that several CYPs and/or non-enzymatic reaction are involved in the formation of GSH adducts from 1",6-dihydroxy BBR. 1'-hydroxybenzbromarone 42-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 27934636-4 2017 The RAF value for CYP3A4 or CYP2C9 derived from a particular P450-selective probe reaction was applied to calculate RAF-scaled CLint for other probe reactions of the same P450 isoform in a crossover manner and compared with the measured HLM CLint When RAF derived from midazolam or nifedipine was used for CYP3A4, the ICR for testosterone 6beta-hydroxylation was 31 and 25, respectively, suggesting significantly diverse interactions of CYP3A4 probes with the testing systems. Nifedipine 282-292 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 27934636-4 2017 The RAF value for CYP3A4 or CYP2C9 derived from a particular P450-selective probe reaction was applied to calculate RAF-scaled CLint for other probe reactions of the same P450 isoform in a crossover manner and compared with the measured HLM CLint When RAF derived from midazolam or nifedipine was used for CYP3A4, the ICR for testosterone 6beta-hydroxylation was 31 and 25, respectively, suggesting significantly diverse interactions of CYP3A4 probes with the testing systems. testosterone 6beta 326-344 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 27934636-6 2017 In addition, these RAF values were applied to losartan and meloxicam, whose metabolism is mostly CYP2C9 mediated. Losartan 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 27934636-6 2017 In addition, these RAF values were applied to losartan and meloxicam, whose metabolism is mostly CYP2C9 mediated. Meloxicam 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 27934636-7 2017 Only using the RAF derived from testosterone for CYP3A4 produced the expected CYP2C9 contribution of 72%-87% and 47%-69% for metabolism of losartan and meloxicam, respectively. Testosterone 32-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27934636-7 2017 Only using the RAF derived from testosterone for CYP3A4 produced the expected CYP2C9 contribution of 72%-87% and 47%-69% for metabolism of losartan and meloxicam, respectively. Losartan 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27117036-0 2017 Warfarin dose requirements with different genotypes of CYP2C9 and VKORC1 for patients with atrial fibrillation and valve replacement. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 27117036-1 2017 AIMS: To investigate whether genetic variants of CYP2C9 and VKORC1 have different effects on the dose of warfarin in 180 Han Chinese patients who were recruited from the Fu Wai Hospital. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 27117036-3 2017 METHODS: DNA was isolated and genotyped using a Warfarin dosage Prediction Kit for single nucleotide polymorphisms (SNPs) of CYP2C9 and VKORC1. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 27117036-4 2017 RESULTS: The VKORC1 and CYP2C9*3 polymorphisms are significantly associated with warfarin maintenance dosages. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 27117036-8 2017 The mean warfarin daily dose requirements in the genotypes of VKORC1 and CYP2C9 were not dependent on the medical indication(s) present. Warfarin 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 27117036-9 2017 CONCLUSIONS: Genetic variants of CYP2C9, VKORC1, and age are significant determinants of the maintenance dose of warfarin. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 27878474-0 2017 Quantitative Assessment of CYP2C9 Genetic Polymorphisms Effect on the Oral Clearance of S-Warfarin in Healthy Subjects. Warfarin 88-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 27878474-1 2017 BACKGROUND: Genetic polymorphisms in CYP2C9 account for 10-20% of the variability in warfarin dose requirement. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 27878474-2 2017 As such CYP2C9 genetic polymorphisms are commonly included in algorithms aimed to optimize warfarin therapy as a way to account for variability in warfarin responsiveness that is due to altered pharmacokinetics. Warfarin 91-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 27878474-2 2017 As such CYP2C9 genetic polymorphisms are commonly included in algorithms aimed to optimize warfarin therapy as a way to account for variability in warfarin responsiveness that is due to altered pharmacokinetics. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 27878474-9 2017 CONCLUSIONS: The current study provides an estimate concerning the effect of CYP2C9 polymorphisms on S-warfarin pharmacokinetics among healthy subjects. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 27052193-12 2017 The results suggest that the altered pharmacokinetics of DIC might be attributed to PIP mediated inhibition of CYP2C9 enzyme, which indicates the clinically significant interaction present between DIC and PIP. piperine 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 28867723-0 2017 The Inhibitory Effect of Telmisartan on the Metabolism of Arachidonic Acid by CYP2C9 and CYP2C8: An in Vitro Study. Telmisartan 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27990512-5 2017 A multiplex allele-specific PCR reaction was used to generate products corresponding to 3 genetic variants associated with warfarin sensitivity [CYP2C9 *2, CYP2C9 *3, and VKORC1 (1173C>T)] and an internal control product. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 27990512-5 2017 A multiplex allele-specific PCR reaction was used to generate products corresponding to 3 genetic variants associated with warfarin sensitivity [CYP2C9 *2, CYP2C9 *3, and VKORC1 (1173C>T)] and an internal control product. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 28867723-0 2017 The Inhibitory Effect of Telmisartan on the Metabolism of Arachidonic Acid by CYP2C9 and CYP2C8: An in Vitro Study. Arachidonic Acid 58-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 28867723-7 2017 Dixon and Lineweaver-Burk plots indicated that telmisartan is a mixed inhibitor of both CYP2C9 and CYP2C8; telmisartan did not show a time-dependent inhibition toward these CYP enzymes. Telmisartan 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 28867723-4 2017 We recently demonstrated that telmisartan, one of angiotensin II receptor blockers, inhibits AA metabolism by CYP enzymes, including CYP2C8, CYP2C9, and CYP2J2. Telmisartan 30-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 28867723-8 2017 Based on the CRs, both CYP2C9 and CYP2C8 are the key enzymes in the metabolism of AA in the human liver. Chromium 13-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 28674251-0 2017 Co-administration of Fluvastatin and CYP3A4 and CYP2C8 Inhibitors May Increase the Exposure to Fluvastatin in Carriers of CYP2C9 Genetic Variants. Fluvastatin 21-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 28867723-5 2017 We conducted studies of AA metabolism using recombinant CYP enzymes to estimate the inhibition constant and the type of inhibition by telmisartan of CYP2C9 and CYP2C8. Telmisartan 134-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 28674251-0 2017 Co-administration of Fluvastatin and CYP3A4 and CYP2C8 Inhibitors May Increase the Exposure to Fluvastatin in Carriers of CYP2C9 Genetic Variants. Fluvastatin 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 27925230-3 2017 Genetic polymorphisms for cytochrome CYP2C9 and VKORC1 explain some of the variability, especially in warfarin and acenocoumarol responses. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 28674251-1 2017 Fluvastatin, which is one of the hydroxymethylglutaryl-CoA (HMG-CoA) reductase inhibitors (statins), is primarily metabolized by CYP2C9 and to a lesser extent by CYP3A4 and CYP2C8. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-4 2017 Since CYP2C9 is a polymorphic enzyme, we investigated the effect of DDI via CYP2C9, CYP3A4, and CYP2C8 on fluvastatin pharmacokinetics by using a validated prediction method in relation to CYP2C9 variants. Fluvastatin 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 6-12 28674251-4 2017 Since CYP2C9 is a polymorphic enzyme, we investigated the effect of DDI via CYP2C9, CYP3A4, and CYP2C8 on fluvastatin pharmacokinetics by using a validated prediction method in relation to CYP2C9 variants. Fluvastatin 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 28674251-4 2017 Since CYP2C9 is a polymorphic enzyme, we investigated the effect of DDI via CYP2C9, CYP3A4, and CYP2C8 on fluvastatin pharmacokinetics by using a validated prediction method in relation to CYP2C9 variants. Fluvastatin 106-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 116-122 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-5 2017 The predicted increases in the area under the concentration-time curve (AUC) ratios of fluvastatin in carriers with CYP2C9*1/*2, CYP2C9*1/*3, CYP2C9*2/*2, CYP2C9*2/*3, and CYP2C9*3/*3 versus that found in carriers with CYP2C9*1/*1 were 1.16, 1.35, 1.37, 1.65, and 2.06, respectively. Fluvastatin 87-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 28674251-6 2017 Our in silico model predicted that administration of fluvastatin in conjunction with the potent inhibitors that completely inhibited CYP3A4 and CYP2C8 in carriers with the CYP2C9*3/*3 variant would cause a 3.23- and 2.60-fold increase in the AUC ratios, respectively, when compared to that for the carriers with the CYP2C9*1/*1 taking fluvastatin alone. Fluvastatin 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 28674251-6 2017 Our in silico model predicted that administration of fluvastatin in conjunction with the potent inhibitors that completely inhibited CYP3A4 and CYP2C8 in carriers with the CYP2C9*3/*3 variant would cause a 3.23- and 2.60-fold increase in the AUC ratios, respectively, when compared to that for the carriers with the CYP2C9*1/*1 taking fluvastatin alone. Fluvastatin 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 316-322 28674251-6 2017 Our in silico model predicted that administration of fluvastatin in conjunction with the potent inhibitors that completely inhibited CYP3A4 and CYP2C8 in carriers with the CYP2C9*3/*3 variant would cause a 3.23- and 2.60-fold increase in the AUC ratios, respectively, when compared to that for the carriers with the CYP2C9*1/*1 taking fluvastatin alone. Fluvastatin 335-346 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 172-178 27925230-3 2017 Genetic polymorphisms for cytochrome CYP2C9 and VKORC1 explain some of the variability, especially in warfarin and acenocoumarol responses. Acenocoumarol 115-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 29472822-6 2017 This case report addresses the influence of CYP2C9 genetic polymorphism (a single nucleotide polymorphism) on phenytoin drug metabolism, thereby causing DRESS syndrome. Phenytoin 110-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 28867752-3 2017 Single nucleotide polymorphisms (SNPs) in CYP2C9 and VKORC1 play a role in the pharmacokinetics and dynamics of warfarin and acenocoumarol and they determine the efficacy of treatment by controlling drug clearance in treated individuals. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 28867752-3 2017 Single nucleotide polymorphisms (SNPs) in CYP2C9 and VKORC1 play a role in the pharmacokinetics and dynamics of warfarin and acenocoumarol and they determine the efficacy of treatment by controlling drug clearance in treated individuals. Acenocoumarol 125-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 28867752-0 2017 Genotyping of CYP2C9 and VKORC1 polymorphisms predicts south Indian patients with deep vein thrombosis as fast metabolizers of warfarin/acenocoumarin. Warfarin 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 28867752-8 2017 None of the patients tested positive for poor drug metabolizing genotypes of the CYP2C9 gene and only 1.6% of the south Indian population was sensitive to warfarin treatment. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 28867752-0 2017 Genotyping of CYP2C9 and VKORC1 polymorphisms predicts south Indian patients with deep vein thrombosis as fast metabolizers of warfarin/acenocoumarin. Acenocoumarol 136-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 27638959-7 2016 CYP2C9-humanized mice have similar bile acid metabolites as the Cyp2c-null mice, indicating that human CYP2C9 does not oxidize CDCA and UDCA, thus explaining the species differences in production of MCA. Bile Acids and Salts 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27889507-0 2017 CYP2C9 Genotypes Modify Benzodiazepine-Related Fall Risk: Original Results From Three Studies With Meta-Analysis. Benzodiazepines 24-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27889507-1 2017 OBJECTIVE: To investigate whether the CYP2C9*2 and *3 variants modify benzodiazepine-related fall risk. Benzodiazepines 70-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 27889507-13 2017 CYP2C9*2 or *3 allele variants modified benzodiazepine-related fall risk. Benzodiazepines 40-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27889507-14 2017 Compared to nonusers, those carrying a CYP2C9*2 or *3 allele and using benzodiazepines had a 45% increased fall risk (HR, 1.45 95% CI, 1.21; 1.73), whereas CYP2C9*1 homozygotes using benzodiazepines had no increased fall risk (HR, 1.14; 95% CI, 0.90; 1.45). Benzodiazepines 183-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 27889507-15 2017 Within benzodiazepine users, having a CYP2C9*2 or *3 allele was associated with an increased fall risk (HR, 1.35; 95% CI, 1.06; 1.72). Benzodiazepines 7-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 27889507-17 2017 CONCLUSIONS: CYP2C9*2 and *3 allele variants modify benzodiazepine-related fall risk. Benzodiazepines 52-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 27889507-20 2017 However, because the exact role of CYP2C9 in benzodiazepine metabolism is still unclear, additional research is warranted. Benzodiazepines 45-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 26454199-3 2017 Warfarin"s more potent enantiomer is primarily metabolized by cytochrome P450 2C9 and has a narrow therapeutic window. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-81 27743182-11 2017 Our study suggests that polymorphic variants of VKORC1 and CYP2C9 affect warfarin dose independently, whereas CYP2C19 did not contribute to warfarin therapy. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 28079798-8 2017 In the dosing algorithm we developed, we confirmed the strongest effects of VKORC1, CYP2C9 on warfarin dosing. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 27967328-4 2017 RESULTS: CYP2C9 and VKORC1 genotypes, age and weight affected acenocoumarol dose and predicted 53.9% of its variability. Acenocoumarol 62-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 27992547-2 2016 Pharmacogenomics studies have shown that variants in CYP2C9 and VKORC1 genes are strongly and consistently associated with warfarin dose variability. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 27992547-10 2016 Variants in the VKORC1 and CYP2C9 were the most significant predictors of warfarin dose in all populations. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 27992547-12 2016 CONCLUSION: Variants of VKORC1 and CYP2C9 are the strongest predictors of warfarin dose variability among the different populations from MENA. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 28302415-13 2017 Patients with CYP2C9 *1/*3 genotype were thrice more likely to have toxic plasma concentrations of phenytoin as compared to those with wild-type genotype (*1/*1) (Adjusted odds ratio - 3.36; 95% C.I. Phenytoin 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 27743182-6 2017 Patient with VKORC1-1639 G > A, CYP2C9*2 and CYP2C9*3 genetic variants need significant lower warfarin dose than patient with wild type allele of VKORC1 1639 G or CYP2C9*1. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 27743182-6 2017 Patient with VKORC1-1639 G > A, CYP2C9*2 and CYP2C9*3 genetic variants need significant lower warfarin dose than patient with wild type allele of VKORC1 1639 G or CYP2C9*1. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 27743182-6 2017 Patient with VKORC1-1639 G > A, CYP2C9*2 and CYP2C9*3 genetic variants need significant lower warfarin dose than patient with wild type allele of VKORC1 1639 G or CYP2C9*1. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 29457079-14 2017 When warfarin and methadone are used together, we have to consider their interaction by comparing the competitive inhibition of CYP2C9 to the induction of CYP3A4 by methadone, because CYP3A4 metabolize various drugs including oxycodone. Warfarin 5-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 29457079-14 2017 When warfarin and methadone are used together, we have to consider their interaction by comparing the competitive inhibition of CYP2C9 to the induction of CYP3A4 by methadone, because CYP3A4 metabolize various drugs including oxycodone. Methadone 18-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 29457079-14 2017 When warfarin and methadone are used together, we have to consider their interaction by comparing the competitive inhibition of CYP2C9 to the induction of CYP3A4 by methadone, because CYP3A4 metabolize various drugs including oxycodone. Oxycodone 226-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 26644206-11 2017 Our study provided another kind of evidence that VKORC1 rs7294, CYP2C9 rs1057910, CYP4F2 rs2108622 and ORM1 rs17650 affected the action of warfarin in different styles. Warfarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 27839691-2 2016 Aceclofenac is converted to 4"-hydroxyaceclofenac and diclofenac via CYP2C9-mediated hydroxylation and hydrolysis, respectively. aceclofenac 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 27839691-2 2016 Aceclofenac is converted to 4"-hydroxyaceclofenac and diclofenac via CYP2C9-mediated hydroxylation and hydrolysis, respectively. 4'-hydroxyaceclofenac 28-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 27839691-2 2016 Aceclofenac is converted to 4"-hydroxyaceclofenac and diclofenac via CYP2C9-mediated hydroxylation and hydrolysis, respectively. Diclofenac 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 27839691-3 2016 CYP2C9 also mediates the hydroxylation of diclofenac to yield 4"-hydroxydiclofenac and the hydrolysis of 4"-hydroxyaceclofenac to 4"-hydroxydiclofenac. Diclofenac 42-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27839691-3 2016 CYP2C9 also mediates the hydroxylation of diclofenac to yield 4"-hydroxydiclofenac and the hydrolysis of 4"-hydroxyaceclofenac to 4"-hydroxydiclofenac. 4'-hydroxydiclofenac 62-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27839691-3 2016 CYP2C9 also mediates the hydroxylation of diclofenac to yield 4"-hydroxydiclofenac and the hydrolysis of 4"-hydroxyaceclofenac to 4"-hydroxydiclofenac. 4'-hydroxyaceclofenac 105-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27839691-3 2016 CYP2C9 also mediates the hydroxylation of diclofenac to yield 4"-hydroxydiclofenac and the hydrolysis of 4"-hydroxyaceclofenac to 4"-hydroxydiclofenac. 4'-hydroxydiclofenac 130-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27885968-1 2016 AIM: To evaluate a possible role of CYP2C9 genotyping for sulfonylureas (SUs) prescription in Russia. Sulfonylurea Compounds 73-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 27198207-0 2016 Multigene and Drug Interaction Approach for Tamoxifen Metabolite Patterns Reveals Possible Involvement of CYP2C9, CYP2C19, and ABCB1. Tamoxifen 44-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 27198207-9 2016 Lower concentrations of endoxifen and endoxifen/4-hydroxytamoxifen ratios were seen with impaired CYP2C9 (P = .05 and P = .03, respectively) if patients had the same CYP2D6 phenotype and were not taking a CYP2D6 or CYP2C19 inhibitor. 4-hydroxy-N-desmethyltamoxifen 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 27198207-9 2016 Lower concentrations of endoxifen and endoxifen/4-hydroxytamoxifen ratios were seen with impaired CYP2C9 (P = .05 and P = .03, respectively) if patients had the same CYP2D6 phenotype and were not taking a CYP2D6 or CYP2C19 inhibitor. 4-hydroxy-N-desmethyltamoxifen 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 27198207-9 2016 Lower concentrations of endoxifen and endoxifen/4-hydroxytamoxifen ratios were seen with impaired CYP2C9 (P = .05 and P = .03, respectively) if patients had the same CYP2D6 phenotype and were not taking a CYP2D6 or CYP2C19 inhibitor. hydroxytamoxifen 48-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 27198207-13 2016 Thus, impairment in CYP2C9, CYP2C19, or ABCB1 contributes to a lower steady-state endoxifen concentration at the dose studied. 4-hydroxy-N-desmethyltamoxifen 82-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 27885968-0 2016 Possible approaches to CYP2C9-guided prescription of sulfonylureas in Russia. Sulfonylurea Compounds 53-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 27885968-2 2016 MATERIALS & METHODS: We have collected the current data on correlation between SUs pharmacodynamics and CYP2C9 polymorphisms. Adenosine Monophosphate 11-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 27885968-1 2016 AIM: To evaluate a possible role of CYP2C9 genotyping for sulfonylureas (SUs) prescription in Russia. Sulfonylurea Compounds 58-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 27885968-2 2016 MATERIALS & METHODS: We have collected the current data on correlation between SUs pharmacodynamics and CYP2C9 polymorphisms. Sulfonylurea Compounds 83-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 27885968-6 2016 CONCLUSION: Considering the received data and the existed knowledge on CYP2C9 influence on SUs pharmacokinetics and pharmacodynamics, we propose a possible approach to CYP2C9-guided SUs prescription for Russians. Sulfonylurea Compounds 91-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 27885968-6 2016 CONCLUSION: Considering the received data and the existed knowledge on CYP2C9 influence on SUs pharmacokinetics and pharmacodynamics, we propose a possible approach to CYP2C9-guided SUs prescription for Russians. Sulfonylurea Compounds 91-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 27885968-6 2016 CONCLUSION: Considering the received data and the existed knowledge on CYP2C9 influence on SUs pharmacokinetics and pharmacodynamics, we propose a possible approach to CYP2C9-guided SUs prescription for Russians. Sulfonylurea Compounds 182-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 27885968-6 2016 CONCLUSION: Considering the received data and the existed knowledge on CYP2C9 influence on SUs pharmacokinetics and pharmacodynamics, we propose a possible approach to CYP2C9-guided SUs prescription for Russians. Sulfonylurea Compounds 182-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 25719441-2 2016 Omeprazole is an acid suppressant and CYP2C9, CYP3A4, and CYP2C19 substrate and inhibitor, as well as inhibitor of transporters (like P-gp). Omeprazole 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 25830869-5 2016 Increased warfarin sensitivity was associated with increased age; CYP2C9 genotypes 2/3, 1/3, and 3/3; VKORC1 genotypes AA and AG; and amiodarone use. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 25830869-6 2016 Decreased warfarin sensitivity (ie, weekly warfarin dose of >21 mg) was associated with increased height, increased weight, having diabetes mellitus, VKORC1 genotype GG, and CYP2C9 genotype 1/1. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 25830869-6 2016 Decreased warfarin sensitivity (ie, weekly warfarin dose of >21 mg) was associated with increased height, increased weight, having diabetes mellitus, VKORC1 genotype GG, and CYP2C9 genotype 1/1. Warfarin 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 177-183 27488389-5 2016 RESULTS: A predictive model of warfarin maintenance dose was established and indicated that age, gender, body surface area, aspirin use, CYP2C9*3, CYP4F2 rs2108622, VKORC1 -1639C>T, and POR*37 831-35C>T accounted for 42.4 % of dose variance in patients undergoing anticoagulant treatment. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 27581200-10 2016 CONCLUSION: The SNP C609T within NQO1 and haplotypes of CYP2C9 (1*2 or 1*3) are independently associated to bleeding complications of warfarin at normal INR. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 27561267-8 2016 A variant genotype of CYP2C9 (i.e., *1/*2 and *1/*3) explained also a part of the remaining variability on Gb clearance. Glyburide 107-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 27393744-0 2016 Implementing Algorithm-Guided Warfarin Dosing in an Ethnically Diverse Patient Population Using Electronic Health Records and Preemptive CYP2C9 and VKORC1 Genetic Testing. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 27632229-3 2016 We genotyped 12 single nucleotide polymorphisms in genes (CYP2C9, VKORC1, CYP4F2, GGCX, EPHX1, CALU) associated with altered warfarin dose requirements and tested their associations with international normalized ratio variability (INRVAR) and percent time in therapeutic range in European-Americans and African-Americans. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 27182616-1 2016 OBJECTIVE: The aim was to investigate the frequency of genetic polymorphisms of cytochrome P4502C9 (CYP2C9) and vitamin K epoxide reductase complex subunit1 (VKORC1) and determine the effect of these polymorphisms on warfarin dose requirements in pediatric patients. Warfarin 217-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-98 27182616-1 2016 OBJECTIVE: The aim was to investigate the frequency of genetic polymorphisms of cytochrome P4502C9 (CYP2C9) and vitamin K epoxide reductase complex subunit1 (VKORC1) and determine the effect of these polymorphisms on warfarin dose requirements in pediatric patients. Warfarin 217-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 27182616-5 2016 The ideal warfarin dose was calculated according to the patient"s age, height, and the presence of CYP2C9*2, CYP2C9*3, and VKORC1 genetic polymorphisms. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 27182616-5 2016 The ideal warfarin dose was calculated according to the patient"s age, height, and the presence of CYP2C9*2, CYP2C9*3, and VKORC1 genetic polymorphisms. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 27258224-0 2016 Effect of CYP2C9 and VKORC1 Gene Variants on Warfarin Response in Patients with Continuous-Flow Left Ventricular Assist Devices. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 30645821-14 2016 Ruxolitinib is mainly metabolised by cytochrome P450 isoenzymes CYP3A4 and CYP2C9, creating a risk of multiple drug interactions. ruxolitinib 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 26750984-10 2016 Glycyrol decreased CYP2C9-catalyzed diclofenac 4"-hydroxylation activity with IC50 values of 0.67 muM in human recombinant cDNA-expressed CYP2C9. glycyrol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 26750984-10 2016 Glycyrol decreased CYP2C9-catalyzed diclofenac 4"-hydroxylation activity with IC50 values of 0.67 muM in human recombinant cDNA-expressed CYP2C9. glycyrol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 26750984-10 2016 Glycyrol decreased CYP2C9-catalyzed diclofenac 4"-hydroxylation activity with IC50 values of 0.67 muM in human recombinant cDNA-expressed CYP2C9. Diclofenac 36-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 27606428-13 2016 The pharmacogenetics studies confirmed that patients carrying the VKORC1 variant-allele required lower warfarin maintenance dosage and that the combination of VKORC1 and CYP2C9 yielded a warfarin responsive index (WRI) inversely related to the number variant alleles. Warfarin 187-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 26750984-7 2016 Glycyrol strongly inhibited CYP1A-mediated phenacetin O-deethylation and CYP2C9-mediated diclofenac 4"-hydroxylation in HLMs, which were the result of competitive inhibition as revealed by a Dixon plot. glycyrol 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 26750984-7 2016 Glycyrol strongly inhibited CYP1A-mediated phenacetin O-deethylation and CYP2C9-mediated diclofenac 4"-hydroxylation in HLMs, which were the result of competitive inhibition as revealed by a Dixon plot. Diclofenac 89-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 27474842-3 2016 The aim of this study is to describe the distribution of CYP2C9*2, CYP2C9*3, CYP3A4*22 and CYP3A5*3 defective alleles, according to losartan tolerance in paediatric Marfan patients. Losartan 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27474842-3 2016 The aim of this study is to describe the distribution of CYP2C9*2, CYP2C9*3, CYP3A4*22 and CYP3A5*3 defective alleles, according to losartan tolerance in paediatric Marfan patients. Losartan 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 27155586-3 2016 The objective of the current study was, in comparison to the standard-therapy method, to investigate the potential of rivaroxaban to improve the treatment of patients with PE, and to reduce hemorrhage in the standard-therapy group through adjusting the dose of warfarin by CYP2C9 and VKORC1 genotypes. Rivaroxaban 118-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 273-279 27353638-0 2016 Impact of age, gender and CYP2C9/2C19 genotypes on dose-adjusted steady-state serum concentrations of valproic acid-a large-scale study based on naturalistic therapeutic drug monitoring data. Valproic Acid 102-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 27390048-4 2016 METHODS: We developed the phenytoin-capecitabine interaction model on the assumption that fluorouracil (5-FU) inhibits cytochrome P450 (CYP) 2C9 synthesis in a concentration- dependent manner. Phenytoin 26-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-144 27390048-4 2016 METHODS: We developed the phenytoin-capecitabine interaction model on the assumption that fluorouracil (5-FU) inhibits cytochrome P450 (CYP) 2C9 synthesis in a concentration- dependent manner. Capecitabine 36-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-144 27390048-4 2016 METHODS: We developed the phenytoin-capecitabine interaction model on the assumption that fluorouracil (5-FU) inhibits cytochrome P450 (CYP) 2C9 synthesis in a concentration- dependent manner. Fluorouracil 90-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-144 27390048-4 2016 METHODS: We developed the phenytoin-capecitabine interaction model on the assumption that fluorouracil (5-FU) inhibits cytochrome P450 (CYP) 2C9 synthesis in a concentration- dependent manner. Fluorouracil 104-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-144 27390048-9 2016 The 50% inhibitory 5-FU concentration for CYP2C9 synthesis and the degradation rate constant of CYP2C9 were estimated to be 0.00310 ng/mL and 0.0768 day-1, respectively. Fluorouracil 19-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 27390048-9 2016 The 50% inhibitory 5-FU concentration for CYP2C9 synthesis and the degradation rate constant of CYP2C9 were estimated to be 0.00310 ng/mL and 0.0768 day-1, respectively. Fluorouracil 19-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 27306545-12 2016 The removal of patient covariates, even highly influential covariates such as CYP2C9 genotype for voriconazole, had no clinical impact. Voriconazole 98-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27155586-14 2016 Implication of CYP2C9 and VKORC1 genotypes in determining dose of warfarin, however, remains to be further examined in larger cohort studies. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 27485379-0 2016 Commentary on clinical significance of CYP2C9-status-guided valproic acid therapy in children. Valproic Acid 60-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)imidazo(2,1-b)(1,3)thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime 63-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 237-243 27197997-6 2016 Activation of CAR and PXR in cells treated with a high dose of CITCO [6-(4-chlorophenyl)-imidazo(2,1-b)thiazole-5-carbaldehyde] or cotreated with rifampicin and tetracycline resulted in synergistic enhancement of CYP3A4, but not CYP2B6, CYP2C9, or UGT1A1, mRNA expression in HepTR/hCAR/hPXR cells. 6-(4-chlorophenyl)-imidazo(2,1-b)thiazole 70-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 237-243 27485380-0 2016 In response: Commentary on clinical significance of CYP2C9-status-guided valproic acid therapy in children. Valproic Acid 73-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Warfarin 187-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Voriconazole 197-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27191765-1 2016 OBJECTIVE: Genetic polymorphisms of the P450 2C9 enzyme (CYP2C9), CYP2C19 and CYP3A5 gene are known to affect the metabolism of many drugs applied in liver transplant recipients, such as warfarin, voriconazole, and tacrolimus. Tacrolimus 215-225 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27262824-2 2016 There are some reports of warfarin dose regimens correlating with single nucleotide polymorphisms (SNP) for CYP2C9, VKORC1 and other genes in adults. Warfarin 26-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 27462768-4 2016 CYP2C9*2 and CYP2C9*3 mediated warfarin sensitivity was observed when vitamin K is in high and low tertiles, respectively. Vitamin K 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 27142473-6 2016 For the vitamin K antagonists, the evidence of the association between variants in VKORC1 and CYP2C9 and the dose is clear. Vitamin K 8-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 27462768-4 2016 CYP2C9*2 and CYP2C9*3 mediated warfarin sensitivity was observed when vitamin K is in high and low tertiles, respectively. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27398225-1 2016 BACKGROUND: Regorafenib and its metabolites may inhibit the activities of several CYP or UDP-glucuronosyltransferase isoforms, including that of CYP2C9. regorafenib 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 27462768-4 2016 CYP2C9*2 and CYP2C9*3 mediated warfarin sensitivity was observed when vitamin K is in high and low tertiles, respectively. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 27462768-4 2016 CYP2C9*2 and CYP2C9*3 mediated warfarin sensitivity was observed when vitamin K is in high and low tertiles, respectively. Vitamin K 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27161631-5 2016 Furthermore, VT-1129 weakly inhibited human CYP2C9, CYP2C19, and CYP3A4, suggesting a low drug-drug interaction potential. VT-1129 13-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 27617219-2 2016 Single nucleotide polymorphisms (SNPs) of CYP2C9, CYP4F2, VKORC1 1173 and VKORC1-1639 influence warfarin maintenance dosage. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 27398225-2 2016 Therefore, pharmacological agents that are CYP2C9 substrates may show elevated circulating levels and enhanced drug efficacy when concurrently used with regorafenib. regorafenib 153-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 27398225-3 2016 Previous studies showed that the area under the plasma concentration-time curve of warfarin, which is the substrate for CYP2C9, increased upon co-administration of regorafenib. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 27398225-3 2016 Previous studies showed that the area under the plasma concentration-time curve of warfarin, which is the substrate for CYP2C9, increased upon co-administration of regorafenib. regorafenib 164-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 27377818-9 2016 CYP2C9*3 and CYP2C9*13 alleles significantly affected the plasma concentrations of zafirlukast. zafirlukast 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 27377818-9 2016 CYP2C9*3 and CYP2C9*13 alleles significantly affected the plasma concentrations of zafirlukast. zafirlukast 83-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 27377818-0 2016 Effects of CYP2C9 genetic polymorphisms on the pharmacokinetics of zafirlukast. zafirlukast 67-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 27302079-5 2016 Here, we have investigated the structural features of CYP2C8, CYP2C9, and CYP2C19 bound with their shared substrate diclofenac to elucidate the underlying molecular mechanism for the substrate regioselectivity of CYP2C subfamily enzymes. Diclofenac 116-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 27377818-2 2016 Zafirlukast is metabolized mainly by CYP3A4 and CYP2C9. zafirlukast 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 27377818-4 2016 A single 20-mg oral dose of zafirlukast was given to 23 Korean male subjects divided into two genotype groups according to CYP2C9 genotypes, CYP2C9EM (n = 11; CYP2C9*1/*1) and CYP2C9IM (n = 12; 9 and 3 carriers of CYP2C9*1/*3 and *1/*13, respectively). zafirlukast 28-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 27377818-6 2016 Compared with the CYP2C9EM group, the Cmax and AUCinf of zafirlukast in the CYP2C9IM group were 1.44- and 1.70-fold higher, respectively (p < 0.01 and p < 0.0001). zafirlukast 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 27377818-6 2016 Compared with the CYP2C9EM group, the Cmax and AUCinf of zafirlukast in the CYP2C9IM group were 1.44- and 1.70-fold higher, respectively (p < 0.01 and p < 0.0001). zafirlukast 57-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 27377818-7 2016 The CL/F of zafirlukast was 42.8 % lower in the CYP2C9IM group compared with the CYP2C9EM group (p < 0.001). Fluorine 7-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 27377818-7 2016 The CL/F of zafirlukast was 42.8 % lower in the CYP2C9IM group compared with the CYP2C9EM group (p < 0.001). zafirlukast 12-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 26581561-5 2016 Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. Amodiaquine 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 26581561-5 2016 Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. montelukast 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 27154258-5 2016 In addition to immune mechanism, metabolism has been found to play a role in the pathogenesis of SCAR, such as recent findings of strong association of CYP2C9*3 with phenytoin-induced SCAR and impaired renal function with allopurinol SCAR. Phenytoin 166-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 27154258-5 2016 In addition to immune mechanism, metabolism has been found to play a role in the pathogenesis of SCAR, such as recent findings of strong association of CYP2C9*3 with phenytoin-induced SCAR and impaired renal function with allopurinol SCAR. Allopurinol 222-233 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 152-158 27163851-0 2016 In vitro metabolism of phenytoin in 36 CYP2C9 variants found in the Chinese population. Phenytoin 23-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 27163851-3 2016 This study aims to assess the catalytic activities of wild-type (CYP2C9*1) and 36 CYP2C9 allelic variants found in the Chinese population toward phenytoin (PHT) in vitro. Phenytoin 145-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 27163851-3 2016 This study aims to assess the catalytic activities of wild-type (CYP2C9*1) and 36 CYP2C9 allelic variants found in the Chinese population toward phenytoin (PHT) in vitro. Phenytoin 156-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 27163851-3 2016 This study aims to assess the catalytic activities of wild-type (CYP2C9*1) and 36 CYP2C9 allelic variants found in the Chinese population toward phenytoin (PHT) in vitro. Phenytoin 145-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 27163851-3 2016 This study aims to assess the catalytic activities of wild-type (CYP2C9*1) and 36 CYP2C9 allelic variants found in the Chinese population toward phenytoin (PHT) in vitro. Phenytoin 156-159 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 27163851-4 2016 Insect microsomes expressing CYP2C9*1 and 36 CYP2C9 variants were incubated with 1-200 muM phenytoin for 30 min at 37 C. Then, these products were extracted and the signal detection was performed by HPLC-MS/MS. Phenytoin 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 27130197-5 2016 The results of the present study show that both formation of 4"-hydroxydiclofenac and it subsequent bioactivation to DF-1",4"-QI is selectively catalyzed by CYP2C9. 4'-hydroxydiclofenac 61-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 27163851-4 2016 Insect microsomes expressing CYP2C9*1 and 36 CYP2C9 variants were incubated with 1-200 muM phenytoin for 30 min at 37 C. Then, these products were extracted and the signal detection was performed by HPLC-MS/MS. Phenytoin 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 27163851-7 2016 The findings suggest that more attention should be paid on subjects carrying these infrequent CYP2C9 alleles when administering phenytoin in clinic. Phenytoin 128-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 27130197-5 2016 The results of the present study show that both formation of 4"-hydroxydiclofenac and it subsequent bioactivation to DF-1",4"-QI is selectively catalyzed by CYP2C9. df-1",4"-qi 117-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 27130197-6 2016 However, the two-step bioactivation to DF-2,5-QI appears to be catalyzed with highest activity by two different CYPs: 5-hydroxylation of DF is predominantly catalyzed by CYP3A4, whereas its subsequent bioactivation to DF-2,5-QI is catalyzed with 14-fold higher intrinsic clearance by CYP2C9. df-2,5-qi 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 27329697-5 2016 In humans, plumbagin was not only a mixed inhibitor of CYP2B6, CYP2C9, CYP2D6, CYP2E1 and CYP3A4, but also a non-competitive inhibitor of CYP1A2, with Ki values no more than 2.16 muM. plumbagin 11-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 26551762-0 2016 The Role of CYP2C8 and CYP2C9 Genotypes in Losartan-Dependent Inhibition of Paclitaxel Metabolism in Human Liver Microsomes. Losartan 43-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 27334984-2 2016 In adults, numerous studies confirmed the robust relationship between warfarin maintenance doses and single nucleotide polymorphisms of cytochrome P450 2C9 (CYP2C9), vitamin K epoxide reductase (VKORC1), and cytochrome P450 4F2 (CYP4F2). Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-155 27334984-2 2016 In adults, numerous studies confirmed the robust relationship between warfarin maintenance doses and single nucleotide polymorphisms of cytochrome P450 2C9 (CYP2C9), vitamin K epoxide reductase (VKORC1), and cytochrome P450 4F2 (CYP4F2). Warfarin 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 27334984-4 2016 The primary objective of the present systematic review and meta-analysis is to assess the effect of genotypes of CYP2C9, VKORC1, and CYP4F2 on warfarin maintenance dose in children. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 27334984-9 2016 DISCUSSION: Our study will provide a comprehensive systematic review and meta-analysis on the potential effects of CYP2C9, VKORC1, or CYP4F2 on the warfarin maintenance dose in children, exploring the feasibility of the development of pharmacogenetic-guided warfarin dosing algorithm for children on oral vitamin K antagonists. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 27296832-1 2016 BACKGROUND: To investigate the associations between atorvastatin-induced liver injury (AILI) and polymorphisms in eight genes possibly involved in the hepatic metabolism (CYP2C9, CYP2C19, CYP3A4, CYP3A5 and UGT1A1) and membrane transport (ABCB1, ABCG2 and SLCO1B1) of atorvastatin, we genotyped 30 AILI and 414 non-AILI patients recruited at BioBank Japan for 15 single nucleotide polymorphisms (SNPs). Atorvastatin 52-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 26551762-0 2016 The Role of CYP2C8 and CYP2C9 Genotypes in Losartan-Dependent Inhibition of Paclitaxel Metabolism in Human Liver Microsomes. Paclitaxel 76-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 26551762-1 2016 The aim of the present study was to further investigate a previously identified metabolic interaction between losartan and paclitaxel, which is one of the marker substrates of CYP2C8, by using human liver microsomes (HLMs) from donors with different CYP2C8 and CYP2C9 genotypes. Losartan 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 261-267 26551762-1 2016 The aim of the present study was to further investigate a previously identified metabolic interaction between losartan and paclitaxel, which is one of the marker substrates of CYP2C8, by using human liver microsomes (HLMs) from donors with different CYP2C8 and CYP2C9 genotypes. Paclitaxel 123-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 261-267 26551762-2 2016 Although CYP2C8 and CYP2C9 exhibit genetic linkage, previous studies have yet to determine whether losartan or its active metabolite, EXP-3174 which is specifically generated by CYP2C9, is responsible for CYP2C8 inhibition. Losartan 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 26551762-6 2016 HLMs with either CYP2C9*2/*2 or CYP2C9*1/*3 exhibited a lower losartan intrinsic clearance (Vmax /Km ) than other HLMs including those with CYP2C9*1/*1 and CYP2C9*1/*2. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 26551762-6 2016 HLMs with either CYP2C9*2/*2 or CYP2C9*1/*3 exhibited a lower losartan intrinsic clearance (Vmax /Km ) than other HLMs including those with CYP2C9*1/*1 and CYP2C9*1/*2. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 26551762-6 2016 HLMs with either CYP2C9*2/*2 or CYP2C9*1/*3 exhibited a lower losartan intrinsic clearance (Vmax /Km ) than other HLMs including those with CYP2C9*1/*1 and CYP2C9*1/*2. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 26551762-6 2016 HLMs with either CYP2C9*2/*2 or CYP2C9*1/*3 exhibited a lower losartan intrinsic clearance (Vmax /Km ) than other HLMs including those with CYP2C9*1/*1 and CYP2C9*1/*2. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 27511999-8 2016 In turn, VKORC1 c.-1639A, CYP2C9*2 and *3 variants were independently associated with actual warfarin dose (P < 0.0001). Warfarin 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 26802434-4 2016 The field of pharmacogenetics is now yielding clinically important results, with three examples outlined: sulphonylurea sensitivity in patients with HNF1A maturity-onset diabetes of the young; sulphonylurea sensitivity in patients with Type 2 diabetes with reduced function alleles at CYP2C9, resulting in reduced metabolism of sulphonylureas; and severe metformin intolerance associated with reduced function organic cation transporter 1 (OCT1) variants, exacerbated by drugs that also inhibit OCT1. Sulfonylurea Compounds 193-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 285-291 27511999-10 2016 Possession of CYP2C9*2 and/or CYP2C9*3 allele variants is associated with lower TTR values and warfarin dose variations in AVR patients, the latter affected also by VKORC1 c.-1693G>A polymorphism. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 27511999-10 2016 Possession of CYP2C9*2 and/or CYP2C9*3 allele variants is associated with lower TTR values and warfarin dose variations in AVR patients, the latter affected also by VKORC1 c.-1693G>A polymorphism. Warfarin 95-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 26122019-4 2016 In multivariate models, CYP2C9 IVS8-109 T was significantly associated with higher PHT plasma concentrations (t(64)=2.27; P=0.03). Phenytoin 83-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 27214159-1 2016 PURPOSE: Some reports have suggested that bucolome, an inhibitor of cytochrome P450 2C9, is useful for decreasing inter-patient variation in warfarin clearance. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-87 26818625-0 2016 S-Warfarin Limited Sampling Models to Estimate Area Under the Concentration Versus Time Curve for Cytochrome P450 2C9 Baseline Activity and After Induction. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-117 26818625-1 2016 BACKGROUND: Phenotyping cytochrome P450 (CYP) 2C9 activity using S-warfarin has routinely required extensive blood sampling over at least 96 hours after dose to estimate the area under the concentration time curve from zero to infinity (AUC). Warfarin 65-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-49 26818625-7 2016 RESULTS: Different results were observed for S-warfarin LSMs in estimating CYP2C9 baseline activity, with most studies resulting in unacceptable bias and precision. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 27179628-0 2016 Frequencies of CYP2C9 polymorphisms in North Indian population and their association with drug levels in children on phenytoin monotherapy. Phenytoin 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 27179628-3 2016 METHODS: We studied 89 epileptic children of North Indian population, randomly selected, to see the genotypic and allelic frequency of CYP2C9 and its association with drug levels on phenytoin monotherapy. Phenytoin 182-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 135-141 27179628-10 2016 CYP2C9*3 allelic group showed significantly higher serum phenytoin levels compared to the wild variants (P = 0.009). Phenytoin 57-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26948084-1 2016 Aprepitant is a known inducer of CYP2C9, the main warfarin-metabolizing enzyme. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 26928377-3 2016 This study investigated the associations between the genetic polymorphisms of HLA class I and CYP2C9 and phenytoin-related SCAR in a Thai population. Phenytoin 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 27298492-2 2016 Considering the paucity of data on the polymorphisms of CYP2C9 in Western Indian population, the present study was conducted to evaluate the prevalence of CYP2C9 polymorphisms (*1, *2 and *3) and correlate it with the activity using flurbiprofen (FLB) as a probe drug. Flurbiprofen 233-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 27298492-2 2016 Considering the paucity of data on the polymorphisms of CYP2C9 in Western Indian population, the present study was conducted to evaluate the prevalence of CYP2C9 polymorphisms (*1, *2 and *3) and correlate it with the activity using flurbiprofen (FLB) as a probe drug. Flurbiprofen 247-250 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 26928377-9 2016 The higher risk of phenytoin-related SJS/TEN was observed in the patients with CYP2C9*3 (odds ratio=4.30, 95% confidence interval=1.41-13.09, P<0.05). Phenytoin 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 26928377-11 2016 The CYP2C9*3 variant was significantly associated with phenytoin-related SJS/TEN, but not DRESS. Phenytoin 55-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 27128896-3 2016 Aschantin potently inhibited CYP2C8-mediated amodiaquine N-de-ethylation, CYP2C9-mediated diclofenac 4"-hydroxylation, CYP2C19-mediated [S]-mephenytoin 4"-hydroxylation, and CYP3A4-mediated midazolam 1"-hydroxylation, with Ki values of 10.2, 3.7, 5.8, and 12.6 microM, respectively. aschantin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 26996562-1 2016 PURPOSE: The genes for cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) have been identified as important genetic determinants of warfarin dosing and have been studied. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-42 26996562-1 2016 PURPOSE: The genes for cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) have been identified as important genetic determinants of warfarin dosing and have been studied. Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 27128896-3 2016 Aschantin potently inhibited CYP2C8-mediated amodiaquine N-de-ethylation, CYP2C9-mediated diclofenac 4"-hydroxylation, CYP2C19-mediated [S]-mephenytoin 4"-hydroxylation, and CYP3A4-mediated midazolam 1"-hydroxylation, with Ki values of 10.2, 3.7, 5.8, and 12.6 microM, respectively. Diclofenac 90-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 27518169-7 2016 Among them, CYP2C9 was most strongly inhibited by (-)-catechin-3-O-gallate (CG) (7.60 microM), and CYP1A2 was most strongly inhibited by EGCG (8.93 microM). catechin gallate 50-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 27199745-0 2016 Functional Characterization of Human CYP2C9 Allelic Variants in COS-7 Cells. carbonyl sulfide 64-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 27199745-2 2016 To evaluate the functional alternations of CYP2C9(*)2, CYP2C9(*)3, CYP2C9(*)8, CYP2C9(*)11 and CYP2C9(*)31, identified in our previous study in Chinese Han population, allelic variants as well as the wild-type CYP2C9 were transiently expressed in COS-7 cells. carbonyl sulfide 247-250 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 27073631-0 2016 Clinical application of a new warfarin-dosing regimen based on the CYP2C9 and VKORC1 genotypes in atrial fibrillation patients. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 27073631-1 2016 The polymorphisms of cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) are important genetic factors for warfarin dose determinations. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-40 27073631-1 2016 The polymorphisms of cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) are important genetic factors for warfarin dose determinations. Warfarin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 27073631-2 2016 The present study aimed to investigate the contribution of the CYP2C9 and VKORC1 genotypes to warfarin dose requirement in atrial fibrillation (AF) patients, and to evaluate the clinical application of a warfarin-dosing algorithm. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 27073631-4 2016 A warfarin-dosing algorithm was developed based on age, height, and the CYP2C9 and VKORC1 genotypes of AF patients. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27073631-5 2016 The results indicated that the mean warfarin daily dose requirement was lower in the CYP2C9*1/*3 genotype compared with those in the homozygous wild-type CYP2C9*1/*1 patients (P<0.05), and was higher in patients with the VKORC1 AG and GG genotypes compared with those with the AA genotype (P<0.05). Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 27073631-6 2016 The multivariate regression model showed that age, height, and the CYP2C9 and VKORC1 genotypes were the best variables for estimating warfarin dose (R2=56.4%). Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 27073631-11 2016 In conclusion, a new warfarin-dosing algorithm was developed based on the CYP2C9 and VKORC1 genotypes, and it can shorten the time elapse from initiation until warfarin maintenance dose in AF patients with warfarin therapy. Warfarin 21-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 27073631-11 2016 In conclusion, a new warfarin-dosing algorithm was developed based on the CYP2C9 and VKORC1 genotypes, and it can shorten the time elapse from initiation until warfarin maintenance dose in AF patients with warfarin therapy. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 27073631-11 2016 In conclusion, a new warfarin-dosing algorithm was developed based on the CYP2C9 and VKORC1 genotypes, and it can shorten the time elapse from initiation until warfarin maintenance dose in AF patients with warfarin therapy. Warfarin 160-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 26878737-4 2016 Genetic variants in CYP2C9 and VKORC1 have been identified and shown to explain some of the variability in warfarin response. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 27518169-7 2016 Among them, CYP2C9 was most strongly inhibited by (-)-catechin-3-O-gallate (CG) (7.60 microM), and CYP1A2 was most strongly inhibited by EGCG (8.93 microM). cg 76-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 27518169-7 2016 Among them, CYP2C9 was most strongly inhibited by (-)-catechin-3-O-gallate (CG) (7.60 microM), and CYP1A2 was most strongly inhibited by EGCG (8.93 microM). epigallocatechin gallate 137-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 27518169-8 2016 Catechin gallate exhibited non-competitive inhibition of CYP2C9, and its Ki value was 9.76 +- 0.47microM. catechin gallate 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27518169-9 2016 The present study is the first to report the inhibitory effect of CG on CYP2C9. cg 66-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27518169-13 2016 The ingestion of beverages containing large amounts of green tea catechins together with drugs that are metabolized by CYP1A2, CYP2C9, and CYP3A4 should be avoided. Catechin 65-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-133 27055637-8 2016 RESULTS: The stable dose of warfarin was tightly associated with factors like age, height, weight, VKORC1 -1639G>A, CYP2C9(*)3, CYP2C9(*)13 and amiodarone usage. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 26940072-0 2016 Association of Genetic Polymorphisms in the VKORC1 and CYP2C9 Genes with Warfarin Dosage in a Group of Kuwaiti Individuals. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 26940072-3 2016 Single nucleotide polymorphisms (SNP) in CYP2C9 and VKORC1 have been shown to significantly affect warfarin dosage toleration and this effect varies among different populations. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 26940072-10 2016 CONCLUSIONS: Our data showed that individuals carrying the wild-type allele of CYP2C9 or VKORC1 rs9923231, rs9934438 or rs2884737 are less sensitive than individuals with the variant alleles of these SNPs and therefore required a higher daily maintenance dose of warfarin. Warfarin 263-271 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 26940072-11 2016 Our study confirms the association between SNPs in CYP2C9 and VKORC1 and warfarin dose tolerance in Kuwaiti patients. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 27080842-4 2016 The best-known examples are AVK and VKORC1-CYP2C9 or clopidogrel and CYP2C19. Clopidogrel 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 26939024-5 2016 To confirm that the emission is from the heme, we destroyed the heme by titration with cumene hydroperoxide and measured the changes in emission upon titration with compounds known to bind to the distal face of the heme in two human cytochrome P450 enzymes, known as CYP3A4 and CYP2C9. Heme 41-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 278-284 26927877-3 2016 Carvedilol is metabolized mainly by glucuronidation and, to a lesser extent, by CYP2D6 to hydroxyphenyl carvedilol (OHC) and by CYP2C9 to O-desmethyl carvedilol (DMC). Carvedilol 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 26927877-3 2016 Carvedilol is metabolized mainly by glucuronidation and, to a lesser extent, by CYP2D6 to hydroxyphenyl carvedilol (OHC) and by CYP2C9 to O-desmethyl carvedilol (DMC). desmethylcarvedilol 138-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 26927877-3 2016 Carvedilol is metabolized mainly by glucuronidation and, to a lesser extent, by CYP2D6 to hydroxyphenyl carvedilol (OHC) and by CYP2C9 to O-desmethyl carvedilol (DMC). desmethylcarvedilol 162-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 27055637-8 2016 RESULTS: The stable dose of warfarin was tightly associated with factors like age, height, weight, VKORC1 -1639G>A, CYP2C9(*)3, CYP2C9(*)13 and amiodarone usage. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 27055637-10 2016 The weights of VKORC1 and CYP2C9 for predicting of warfarin dosage were estimated to more than 50%. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 26877175-2 2016 The CYP2C9 was attached to magnetic SiMAG-carboxyl microparticles using the carbodiimide method. Carbodiimides 76-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 26806573-0 2016 Effects of salvianolic acid B and tanshinone IIA on the pharmacokinetics of losartan in rats by regulating the activities and expression of CYP3A4 and CYP2C9. Losartan 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 26877175-5 2016 The on-line kinetic and inhibition studies of CYP2C9"s reaction with diclofenac as a model substrate and sulfaphenazole as a model inhibitor were conducted in order to demonstrate its practical applicability. Diclofenac 69-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 26877175-5 2016 The on-line kinetic and inhibition studies of CYP2C9"s reaction with diclofenac as a model substrate and sulfaphenazole as a model inhibitor were conducted in order to demonstrate its practical applicability. Sulfaphenazole 105-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 27622442-0 2016 Prediction of sensitivity to warfarin based on VKORC1 and CYP2C9 polymorphisms in patients from different places in Colombia. Warfarin 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 27622442-9 2016 CONCLUSIONS: These results in a mixed population support the prediction of sensitivity to warfarin based on polymorphisms in VKORC1 and CYP2C9 as a valid approach in Colombian patients. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 27092633-3 2016 This review brings an update about how genetic polymorphisms of CYP2C8 and CYP2C9 enzymes affect the disposition and clinical outcomes of ibuprofen and diclofenac, two of the most common pain relievers. Ibuprofen 138-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 27092633-3 2016 This review brings an update about how genetic polymorphisms of CYP2C8 and CYP2C9 enzymes affect the disposition and clinical outcomes of ibuprofen and diclofenac, two of the most common pain relievers. Diclofenac 152-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 27092633-4 2016 The most common side effects associated with the influence of CYP2C8*3 and CYP2C9*2*3 variants on ibuprofen and diclofenac pharmacokinetics are hepatotoxicity and gastrointestinal bleeding. Ibuprofen 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 27092633-4 2016 The most common side effects associated with the influence of CYP2C8*3 and CYP2C9*2*3 variants on ibuprofen and diclofenac pharmacokinetics are hepatotoxicity and gastrointestinal bleeding. Diclofenac 112-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 24978953-7 2016 Our results suggest that the VKORC1-1639 GG and the wild type CYP2C9*1*1genotypes are associated with the high-dose requirement for warfarin therapy, and that VKORC1-1639 GG is responsible for warfarin resistance and failure in Egyptian patients. Warfarin 132-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 24978953-0 2016 VKORC1 and CYP2C9 genotypes in Egyptian patients with warfarin resistance. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24978953-7 2016 Our results suggest that the VKORC1-1639 GG and the wild type CYP2C9*1*1genotypes are associated with the high-dose requirement for warfarin therapy, and that VKORC1-1639 GG is responsible for warfarin resistance and failure in Egyptian patients. Warfarin 193-201 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 24978953-3 2016 The resistance is associated with polymorphisms of the vitamin K epoxide reductase-oxidase complex (VKORC1) and cytochrome P450-2C9 (CYP2C9) genes, which affect warfarin pharmacodynamics and pharmacokinetics, respectively. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-131 27008632-4 2016 METHODS: We genotyped 5 variants of these genes in a total of 747 diabetic patients enrolled in a trial of glibenclamide (CYP2C9, KCNJ11, TCF7L2, KCNQ1 and CDKN2A/2B gene). Glyburide 107-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 24978953-3 2016 The resistance is associated with polymorphisms of the vitamin K epoxide reductase-oxidase complex (VKORC1) and cytochrome P450-2C9 (CYP2C9) genes, which affect warfarin pharmacodynamics and pharmacokinetics, respectively. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 26894931-1 2016 AIM: Correlation of outcomes of cyclophosphamide (CP) therapy in antineutrophil cytoplasmic antibody-associated vasculitis with genotype polymorphisms in prodrug activating cytochrome P450 enzyme genes CYP2C9 and CYP2C19. Cyclophosphamide 32-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 202-208 26267707-2 2016 The dosage of warfarin is strongly affected by genetic variants of CYP2C9 and VKORC1 genes. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 26886310-0 2016 The Effect of Genetic Polymorphism on the Inhibition of Azole Antifungal Agents Against CYP2C9-Mediated Metabolism. Azoles 56-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 26886310-1 2016 We investigated the effect of cytochrome P450 (CYP) 2C9 polymorphism on the inhibition of methylhydroxylation activity of tolbutamide, a typical CYP2C9 substrate, by triazole antifungal agents, fluconazole and voriconazole. Tolbutamide 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-55 26886310-1 2016 We investigated the effect of cytochrome P450 (CYP) 2C9 polymorphism on the inhibition of methylhydroxylation activity of tolbutamide, a typical CYP2C9 substrate, by triazole antifungal agents, fluconazole and voriconazole. Tolbutamide 122-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 26886310-1 2016 We investigated the effect of cytochrome P450 (CYP) 2C9 polymorphism on the inhibition of methylhydroxylation activity of tolbutamide, a typical CYP2C9 substrate, by triazole antifungal agents, fluconazole and voriconazole. Triazoles 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-55 26886310-1 2016 We investigated the effect of cytochrome P450 (CYP) 2C9 polymorphism on the inhibition of methylhydroxylation activity of tolbutamide, a typical CYP2C9 substrate, by triazole antifungal agents, fluconazole and voriconazole. Triazoles 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 145-151 26886310-3 2016 These results suggest that more careful administration of azole antifungals to patients with the CYP2C9*2 allele might be required because of the strong inhibitory effects. Azoles 58-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 26894931-8 2016 CONCLUSION: Patients with variant CYP2C9 are at increased risk for cyclophosphamide-induced leukopenia but may have a better chance to respond to treatment. Cyclophosphamide 67-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 26719074-2 2016 The purpose of this study was to evaluate the pharmacokinetics and pharmacodynamics of warfarin (as a reference drug metabolized by CYP2C9) in the presence and absence of belinostat. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 26633237-8 2016 The results suggest that the altered pharmacokinetics of DIC might be attributed to RSV mediated inhibition of CYP2C9 enzyme. Resveratrol 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 27180666-4 2016 The response to sulfonylurea derivatives was significantly associated with the variants KCNJ11/ABCC8, TCF7L2 and CYP2C9. Sulfonylurea Compounds 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 27026863-0 2016 Metabolic inhibition of meloxicam by specific CYP2C9 inhibitors in Cunninghamella blakesleeana NCIM 687: in silico and in vitro studies. Meloxicam 24-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 27026863-4 2016 The in silico studies were further substantiated by in vitro studies, which indicated fenofibrate to be a potent inhibitor of CYP2C9 enzyme followed by sertraline, clopidogrel and fluvoxamine, respectively. Fenofibrate 86-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 27026863-5 2016 Two-stage fermentation protocol was followed to study metabolism of meloxicam and its inhibition by different CYP2C9 inhibitors. Meloxicam 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 26845774-4 2016 The results showed that CK inhibited the enzyme activities of CYP2C9 and CYP3A4 in the HLMs. ginsenoside M1 24-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 26845774-8 2016 The results suggested that CK was a substrate and also inhibitors for both CYP2C9 and CYP3A4. ginsenoside M1 27-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 26845774-9 2016 Patients using CK in combination with therapeutic drugs that are substrates of CYP2C9 and CYP3A4 for different reasons should be careful, although the inhibiting potency of CK is much poorer than that of enzyme-specific inhibitors. ginsenoside M1 15-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 26658225-5 2016 The EC50 of rifampin for CYP3A4 and CYP2C9 was up to 10-fold lower in HepatoPac than the monocultures. Rifampin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 26751406-1 2016 Genotype-based algorithms that include VKORC1 and CYP2C9 genotypes are less predictive of warfarin dose variability in Africans as opposed to Europeans. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 25476996-0 2016 The role of CYP2C9 genetic polymorphism in carvedilol O-desmethylation in vitro. Carvedilol 43-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25476996-1 2016 We aimed at investigating the role of CYP2C9 in carvedilol O-desmethylation and identifying the effect of 35 CYP2C9 allelic variants we found in Chinese Han population on the in vitro metabolism of carvedilol. Carvedilol 198-208 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 25476996-3 2016 Recombinant CYP2C9 microsomes of distinguished genotypes were used to characterize the corresponding enzyme activity toward carvedilol. Carvedilol 124-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25476996-5 2016 CYP2C9 plays a certain role in carvedilol metabolism. Carvedilol 31-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25476996-6 2016 Compared with wild-type CYP2C9*1, the intrinsic clearance (V max/K m) values of all variants toward carvedilol O-desmethylation were significantly altered. Carvedilol 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 26549688-6 2016 RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. Rifampin 59-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Rifampin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 26568290-6 2016 A two-color multiplex allele-specific PCR assay for detecting the warfarin-related single-nucleotide polymorphisms (SNPs) 6853 (-1639G>A) and 6484 (1173C>T) in the VKORC1 gene and the *3 SNP (1075A>C) in the CYP2C9 gene was developed and used for validation studies. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 26759747-2 2016 Rifampin is a widely used antimicrobial that has major interactions with several medications including warfarin due to its strong P-glycoprotein and liver enzyme inducer activity especially on CYP2C9, CYP3A4, CYP1A2 and CYP2C19. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 26739746-6 2016 Statistical analyses were performed using Plink and R. RESULTS: VKORC1 and CYP2C9 were the major genetic determinants of warfarin MWD and SMWD, with CYP4F2 having a smaller effect. Warfarin 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 26739746-9 2016 Genotypes for VKORC1 and CYP2C9*3, age, height and weight, as well as other clinical factors such as alcohol consumption, loading dose and concomitant drugs were important for the initial INR response to warfarin. Warfarin 204-212 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 27174459-6 2016 RESULTS: The final PPK model equations of oral phenytoin were found to be as follows: for patients with CYP2C9 *1/*1, Vmax=22.66. Phenytoin 47-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 26739746-12 2016 VKORC1 and CYP2C9 genetic polymorphisms are the most important determinants of warfarin dosing, and it is highly unlikely that other common variants of clinical importance influencing warfarin dosage will be found. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 26739746-13 2016 Both VKORC1 and CYP2C9*3 are important determinants of the initial INR response to warfarin. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25998968-0 2016 Phenytoin toxicity in two-month-old Thai infant with CYP2C9 gene polymorphism--A case report. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 25998968-4 2016 The cytochrome P450 super family plays an important role in phenytoin metabolism, especially CYP2C9 and CYP2C19. Phenytoin 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 25998968-5 2016 In this case report, we profiled a two-month-old Thai infant who developed phenytoin toxicity resulting from CYP2C9 gene polymorphism. Phenytoin 75-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 26777610-0 2016 Association of Genetic Polymorphisms of CYP2C9 and VKORC1 with Bleeding Following Warfarin: A Case-Control Study. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 26777610-2 2016 This study aimed to assess the association of CYP2C9 and VKORC1 with the development of bleeding following warfarin. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 26777610-12 2016 CONCLUSION: A significant association between CYP2C9 (*1,*2,*3) genotype and VKORC1 (1639 G>A) haplotype status has been found with increased bleeding tendency to warfarin. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 26496779-7 2016 Phenobarbital is metabolized in the liver by CYP2C9 with minor metabolism by CYP2C19 and CYP2E1. Phenobarbital 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. Losartan 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 26502773-6 2016 In hepatocyte toxicity studies, the cell viability of human hepatocytes decreased after exposure to Ro 47-8634, and the observed hepatotoxicity was diminished by pretreatment with tienilic acid (CYP2C9-specific inactivator). Ticrynafen 180-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. Valsartan 236-245 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. candesartan 247-258 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. Irbesartan 260-270 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. azilsartan 276-286 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. Telmisartan 368-379 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. eprosartan 381-391 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 27216792-6 2016 RESULTS: Based on the literatures published, it has been demonstrated that pharmacokinetic interactions of losartan with other agents are mainly via CYP2C9- and CYP3A4-mediated, the role played by CYP enzyme system in the metabolism of valsartan, candesartan, irbesartan, and azilsartan appears modest, and cytochrome P450 system has no influence on the metabolism of telmisartan, eprosartan, olmesartan. olmesartan 393-403 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 26502773-9 2016 These results suggest that Ro 64-1056 is directly involved in bosentan-induced liver injury partly because CYP2C9 specifically mediates hydroxylation of the t-butyl group of Ro 47-8634. Bosentan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 29787666-3 2016 (Argl44Cys) and CYP2C9*3 (Ile359Leu) genes showed that the dominant genotype of CYP2C9*2 was the honiozygous CC carriership and for CYP2C9*3 it was the prevalence of AA genotype, the incidence of which was close and amounted to 80%. argl44cys 1-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 29787666-3 2016 (Argl44Cys) and CYP2C9*3 (Ile359Leu) genes showed that the dominant genotype of CYP2C9*2 was the honiozygous CC carriership and for CYP2C9*3 it was the prevalence of AA genotype, the incidence of which was close and amounted to 80%. argl44cys 1-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 27006677-4 2016 To evaluate the effects of vinpocetine on the activity of human CYP3A4, CYP2C9, CYP2C19, CYP2D6, and CYP2E1, human liver microsomes were utilized to incubate with the mixed CYPs probe substrates and the target components. vinpocetine 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 26693855-6 2016 RESULTS: Benzbromarone metabolism resulted in inhibition of CYP3A4 but not CYP2C9 in a time-dependent manner. Benzbromarone 9-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 27006677-5 2016 The results indicate that vinpocetine exhibited weak inhibitory effect on the CYP2C9, where the IC50 value is 68.96 muM, whereas the IC50 values for CYP3A4, CYP2C19, CYP2D6, and CYP2E1 were all over range of 100 muM, which showed that vinpocetine had no apparent inhibitory effects on these CYPs. vinpocetine 26-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 27006677-6 2016 In conclusion, the results indicated that drugs metabolized by CYP2C9 coadministrated with vinpocetine may require attention or dose adjustment. vinpocetine 91-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 26355760-7 2016 Linear regression and ANN models identified several predictors of warfarin dose including age, weight, CYP2C9 genotype *1/*1, VKORC1 genotype, rs12777823 genotype, rs2108622 genotype, congestive heart failure, and amiodarone use. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 27698677-7 2016 CYP1A2, CYP2C9, CYP2D6, CYP2E1, CYP3A4, and CYP2C19 contributed differently to the metabolism of morusin. morusin 97-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 26870974-1 2016 The aim of the research was to study the frequency of VKROC1 and CYP2C9 genes different alleles for healthy donors and for patients with thrombosis, in two regions of Georgia - in Samegrelo and in Tbilisi and to reveal the interdependence of the studied genes products in the treatment of thrombosis with warfarin. Warfarin 305-313 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 26870974-3 2016 The protein product of CYP2C9 gene is involved in the metabolism of warfarin. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 28057172-2 2016 METHODS: Aminopyrine N-demethylation and tolbutamide methylhydroxylation by CYP2C8, CYP2C9, and CYP2C19 were determined by the previous reported methods. Aminopyrine 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26655108-3 2016 These two enantiomers show differences in metabolism by CYPs: S-warfarin undergoes 7 hydroxylation by CYP2C9 and R-warfarin by CYP3A4 to form 10 hydroxy warfarin. Warfarin 62-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 26655108-12 2016 In addition, we show inhibition of major metabolic pathways of warfarin by sulfaphenazole and ketoconazole which are known specific inhibitors of CYP2C9 and CYP3A4 respectively. Warfarin 63-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 26655108-12 2016 In addition, we show inhibition of major metabolic pathways of warfarin by sulfaphenazole and ketoconazole which are known specific inhibitors of CYP2C9 and CYP3A4 respectively. Sulfaphenazole 75-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 26655108-12 2016 In addition, we show inhibition of major metabolic pathways of warfarin by sulfaphenazole and ketoconazole which are known specific inhibitors of CYP2C9 and CYP3A4 respectively. Ketoconazole 94-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 28057172-2 2016 METHODS: Aminopyrine N-demethylation and tolbutamide methylhydroxylation by CYP2C8, CYP2C9, and CYP2C19 were determined by the previous reported methods. Tolbutamide 41-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 28057172-6 2016 For tolbutamide methylhydroxylation, the Km and Vmax for CYP2C19 were three and six times higher than the corresponding values for CYP2C9, and the Vmax/Km value for CYP2C19 was twice that for CYP2C9, whereas hydroxylated tolbutamide formed by CYP2C8 was not detected. Tolbutamide 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 28057172-6 2016 For tolbutamide methylhydroxylation, the Km and Vmax for CYP2C19 were three and six times higher than the corresponding values for CYP2C9, and the Vmax/Km value for CYP2C19 was twice that for CYP2C9, whereas hydroxylated tolbutamide formed by CYP2C8 was not detected. Tolbutamide 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 192-198 26569597-4 2016 CYP2C9-based interaction was investigated for both 2-oxo-clopidogrel and glimepiride using HLM and the recombinant CYP2C9 system. 2-oxo-clopidogrel 51-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26569597-4 2016 CYP2C9-based interaction was investigated for both 2-oxo-clopidogrel and glimepiride using HLM and the recombinant CYP2C9 system. glimepiride 73-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26569597-6 2016 For the metabolism of 2-oxo-clopidogrel (the second step of bioactivation), glimepiride demonstrated a relatively strong inhibition against CYP2C9 activity (IC50 12.7 mumol/l). 2-oxo-clopidogrel 22-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 26569597-6 2016 For the metabolism of 2-oxo-clopidogrel (the second step of bioactivation), glimepiride demonstrated a relatively strong inhibition against CYP2C9 activity (IC50 12.7 mumol/l). glimepiride 76-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 26569597-7 2016 In addition, 2-oxo-clopidogrel displayed a moderate inhibitory effect toward the CYP2C9-mediated metabolism of glimepiride. 2-oxo-clopidogrel 13-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 26569597-7 2016 In addition, 2-oxo-clopidogrel displayed a moderate inhibitory effect toward the CYP2C9-mediated metabolism of glimepiride. glimepiride 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 27287328-3 2016 METHODS: Different concentrations of apigenin were added to a 100 mmol/l Tris-HCl reaction mixture containing 2 pmol/ml recombinant human CYP2C9.1, 0.25 mg/ml human liver microsomes or 0.5 mg/ml rat liver microsomes to determine the half maximal inhibition or a half-maximal inhibitory concentration (IC50) on the metabolism of losartan. Apigenin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 27287328-5 2016 RESULTS: The results showed that apigenin has the inhibitory effect on the metabolism of losartan in vitro, the IC50 was 7.61, 4.10 and 11.07 mumol/l on recombinant CYP2C9 microsomes, human liver microsomes and rat liver microsomes, respectively. Apigenin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 27287328-5 2016 RESULTS: The results showed that apigenin has the inhibitory effect on the metabolism of losartan in vitro, the IC50 was 7.61, 4.10 and 11.07 mumol/l on recombinant CYP2C9 microsomes, human liver microsomes and rat liver microsomes, respectively. Losartan 89-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 165-171 27287328-6 2016 Meanwhile, apigenin"s mode of action on human CYP2C9 activity was competitive for the substrate diclofenac. Diclofenac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 26658169-0 2016 Phenytoin neurotoxicity in a child carrying new STXBP1 and CYP2C9 gene mutations. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. Warfarin 2-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. 7-hydroxywarfarin 11-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. 7-hydroxywarfarin 11-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 26666467-7 2016 S-warfarin/7-hydroxywarfarin ratio was found to increase in subjects with CYP2C9*2 and CYP2C9*3 justifying the warfarin sensitivity attributed to these variants. Warfarin 20-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 26409440-5 2015 Significant promotions were also demonstrated in the syntheses of albumin and urea and the activities of phase I (CYP 3A11 and CYP 2C9) and phase II enzymes. Urea 78-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 127-134 30620511-5 2016 Flu-voxamine inhibits CYPlA2 and CYP2C9 activity, and paroxetine inhibits CYP2D6 activity. Fluvoxamine 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 26677906-5 2016 Formation of 5-CA and S-777469 5-hydroxymethyl (5-HM), a precursor metabolite of 5-CA, was catalyzed by CYP2C9. S-777469 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 26677906-5 2016 Formation of 5-CA and S-777469 5-hydroxymethyl (5-HM), a precursor metabolite of 5-CA, was catalyzed by CYP2C9. 5-hydroxymethyl 31-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 26677906-5 2016 Formation of 5-CA and S-777469 5-hydroxymethyl (5-HM), a precursor metabolite of 5-CA, was catalyzed by CYP2C9. 5-hydroxymethyl tolterodine 48-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 26677906-6 2016 Arachidonic acid, alpha-linolenic acid, oleic acid and myristic acid, which have been reported to exist in liver microsomes, inhibited S-777469 oxidation by CYP2C9, but serum albumin enhanced this reactions. Arachidonic Acid 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26677906-6 2016 Arachidonic acid, alpha-linolenic acid, oleic acid and myristic acid, which have been reported to exist in liver microsomes, inhibited S-777469 oxidation by CYP2C9, but serum albumin enhanced this reactions. alpha-Linolenic Acid 18-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26677906-6 2016 Arachidonic acid, alpha-linolenic acid, oleic acid and myristic acid, which have been reported to exist in liver microsomes, inhibited S-777469 oxidation by CYP2C9, but serum albumin enhanced this reactions. Oleic Acid 40-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26677906-6 2016 Arachidonic acid, alpha-linolenic acid, oleic acid and myristic acid, which have been reported to exist in liver microsomes, inhibited S-777469 oxidation by CYP2C9, but serum albumin enhanced this reactions. Myristic Acid 55-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 157-163 26677906-11 2016 CYP2C9 was the enzyme responsible for S-777469 oxidation in human livers. Sulfur 38-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26712778-2 2015 This study evaluated the effect of GA on the activity of five P450(CYP450) cytochrome enzymes: CYP2A6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, in human liver microsomes (HLMs) and recombinant cDNA-expressed enzyme systems using a HPLC-MS/MS CYP-specific probe substrate assay. Glycyrrhetinic Acid 35-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 26712778-6 2015 Moreover, CYP2C9 and CYP2C19 could also be inhibited significantly by GA with IC50 of 42.89 and 40.26 muM in HLMs, respectively. Glycyrrhetinic Acid 70-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 26377995-10 2015 CONCLUSIONS: The genotyping of polymorphic variants in VKORC1 and CYP2C9, together with clinical and demographic parameters, can serve for more precise definition of the individual starting and maintenance doses of coumarin derivatives in each patient. coumarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 26348733-6 2015 Several compounds, including loratadine, showed high selectivity to cynomolgus monkey CYP2C19, and all of these have been reported as human CYP2C19 and/or CYP2C9 substrates. Loratadine 29-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 26223945-7 2015 Similarly, in the subpopulation of AA carriers in the CYP2C9 gene, patients with the T allele required significantly higher doses of warfarin than those with other genotypes of rs887829 (6.5 +- 2.4 vs. 5.3 +- 1.5 mg, P = 0.002). Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 26344676-1 2015 Genetic variation in the pharmacokinetics of metoprolol and torsemide due to polymorphisms in CYP2D6, CYP2C9, and OATP1B1 has been extensively studied. Metoprolol 45-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 26264702-5 2015 RESULTS: In vitro, evacetrapib inhibited all of the major CYPs, with inhibition constants (K(i)) ranging from 0.57 microM (CYP2C9) to 7.6 microM (CYP2C19). evacetrapib 19-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 26344676-1 2015 Genetic variation in the pharmacokinetics of metoprolol and torsemide due to polymorphisms in CYP2D6, CYP2C9, and OATP1B1 has been extensively studied. Torsemide 60-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 26374173-3 2015 Kinetic and inhibition experiments performed at therapeutically relevant concentrations reveal that CYP2C8 and CYP2C9 are the principal enzymes responsible for montelukast 36-hydroxylation to 1,2-diol. montelukast 160-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 26781925-6 2015 This was a prospective hospital based study in which allele and genotypic frequencies of CYP2C9 gene polymorphisms; 430C>T and 1075A>C and VKORC1 gene polymorphisms; 1639G>A, 9041G>A and 6009C>T in 106 alleles of north Indian patients with valve replacement on acenocoumarol were determined and their effect on acenocoumarol dosing was studied. Acenocoumarol 276-289 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 26781925-6 2015 This was a prospective hospital based study in which allele and genotypic frequencies of CYP2C9 gene polymorphisms; 430C>T and 1075A>C and VKORC1 gene polymorphisms; 1639G>A, 9041G>A and 6009C>T in 106 alleles of north Indian patients with valve replacement on acenocoumarol were determined and their effect on acenocoumarol dosing was studied. Acenocoumarol 326-339 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 26781925-8 2015 In 53 patients with valve replacement on acenocoumarol with stable INR, the allele frequency of CYP2C9*2 and CYP2C9*3 gene polymorphisms was 0.05 and 0.17 respectively and that of VKORC1 *2,*3 and *4 gene polymorphisms was 0.15, 0.72 and 0.11 respectively. Acenocoumarol 41-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 26781925-9 2015 The presence of CYP2C9*3 or VKORC1*2 gene polymorphism were associated with decrease in acenocoumarol dose requirements (p values 0.03 and 0.02 respectively).This study confirmed the association of lower mean weekly dosages of acenocoumarol in patients with CYP2C9*3 and VKORC1*2 gene polymorphisms. Acenocoumarol 88-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 26781925-9 2015 The presence of CYP2C9*3 or VKORC1*2 gene polymorphism were associated with decrease in acenocoumarol dose requirements (p values 0.03 and 0.02 respectively).This study confirmed the association of lower mean weekly dosages of acenocoumarol in patients with CYP2C9*3 and VKORC1*2 gene polymorphisms. Acenocoumarol 88-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 258-264 26781925-9 2015 The presence of CYP2C9*3 or VKORC1*2 gene polymorphism were associated with decrease in acenocoumarol dose requirements (p values 0.03 and 0.02 respectively).This study confirmed the association of lower mean weekly dosages of acenocoumarol in patients with CYP2C9*3 and VKORC1*2 gene polymorphisms. Acenocoumarol 227-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 26781925-9 2015 The presence of CYP2C9*3 or VKORC1*2 gene polymorphism were associated with decrease in acenocoumarol dose requirements (p values 0.03 and 0.02 respectively).This study confirmed the association of lower mean weekly dosages of acenocoumarol in patients with CYP2C9*3 and VKORC1*2 gene polymorphisms. Acenocoumarol 227-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 258-264 26409163-8 2015 The in-vitro metabolism assay with the human CYP2C9 isoform led to the formation of THCOH and THCCOOH of Delta(8)-THC and Delta(9)-THC. thcoh 84-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26409163-8 2015 The in-vitro metabolism assay with the human CYP2C9 isoform led to the formation of THCOH and THCCOOH of Delta(8)-THC and Delta(9)-THC. 11-nor-delta(9)-tetrahydrocannabinol-9-carboxylic acid 94-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26409163-8 2015 The in-vitro metabolism assay with the human CYP2C9 isoform led to the formation of THCOH and THCCOOH of Delta(8)-THC and Delta(9)-THC. delta-8-tetrahydrocannabinol 105-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26409163-8 2015 The in-vitro metabolism assay with the human CYP2C9 isoform led to the formation of THCOH and THCCOOH of Delta(8)-THC and Delta(9)-THC. Dronabinol 122-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26374173-3 2015 Kinetic and inhibition experiments performed at therapeutically relevant concentrations reveal that CYP2C8 and CYP2C9 are the principal enzymes responsible for montelukast 36-hydroxylation to 1,2-diol. MLS002693268 192-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 26669712-6 2015 The CYP2C9 phenotype was determined using a single oral dose of 50 mg losartan. Losartan 70-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 26669712-8 2015 The urinary losartan to its metabolite E-3174 metabolic ratio (MR) was used as an index of CYP2C9 metabolic activity. Losartan 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 27047214-0 2015 The influence of VKORC1 and CYP2C9 mutations on warfarin response after total hip and knee arthroplasty. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 26360837-2 2015 The aim of this study was to evaluate the influence of the genetic polymorphisms of CYP2C9, CYP2D6 and CYP3A4 on the pharmacokinetics (PK) of celecoxib and its two main metabolites, hydroxyl-celecoxib and carboxy-celecoxib, in healthy Chinese subjects, based on a bioequivalence study of celecoxib. Celecoxib 191-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26593908-7 2015 Human CYP1A2 followed by CYP2C9, 3A4 and 1A1 were the major enzymes contributing to AAI oxidation in human liver, while CYP2C and 1A were most important in rat liver. aristolochic acid I 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 26360837-2 2015 The aim of this study was to evaluate the influence of the genetic polymorphisms of CYP2C9, CYP2D6 and CYP3A4 on the pharmacokinetics (PK) of celecoxib and its two main metabolites, hydroxyl-celecoxib and carboxy-celecoxib, in healthy Chinese subjects, based on a bioequivalence study of celecoxib. Celecoxib 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26360837-8 2015 Therefore, the influence of the CYP2C9*3 and CYP2D6*10 on the PK of celecoxib and its metabolites in Chinese was studied. Celecoxib 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 26360837-9 2015 Compared with CYP2C9*1/*1 group, pharmacokinetic parameters of celecoxib such as AUC0-48 and Cmax was increased by 90.6% and 45.8%, the t1/2 was extended by 21.8% and the CL/F was decreased by 51.1% in CYP2C9*1/*3 group. Celecoxib 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 26360837-2 2015 The aim of this study was to evaluate the influence of the genetic polymorphisms of CYP2C9, CYP2D6 and CYP3A4 on the pharmacokinetics (PK) of celecoxib and its two main metabolites, hydroxyl-celecoxib and carboxy-celecoxib, in healthy Chinese subjects, based on a bioequivalence study of celecoxib. hydroxyl-celecoxib 182-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26360837-9 2015 Compared with CYP2C9*1/*1 group, pharmacokinetic parameters of celecoxib such as AUC0-48 and Cmax was increased by 90.6% and 45.8%, the t1/2 was extended by 21.8% and the CL/F was decreased by 51.1% in CYP2C9*1/*3 group. Celecoxib 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 202-208 26360837-2 2015 The aim of this study was to evaluate the influence of the genetic polymorphisms of CYP2C9, CYP2D6 and CYP3A4 on the pharmacokinetics (PK) of celecoxib and its two main metabolites, hydroxyl-celecoxib and carboxy-celecoxib, in healthy Chinese subjects, based on a bioequivalence study of celecoxib. carboxy-celecoxib 205-222 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26360837-10 2015 In terms of hydroxy-celecoxib, compared with CYP2C9*1/*1 group, the Cmax was decreased by 17.2%, the t1/2 prolonged 42.1% in CYP2C9*1/*3 group. hydroxy-celecoxib 12-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26360837-10 2015 In terms of hydroxy-celecoxib, compared with CYP2C9*1/*1 group, the Cmax was decreased by 17.2%, the t1/2 prolonged 42.1% in CYP2C9*1/*3 group. hydroxy-celecoxib 12-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 26360837-11 2015 In terms of carboxy-celecoxib, the AUC0-48 was increased by 25.2%, the t1/2 prolonged 16.1% and the CL/F was decreased by 21.2% in CYP2C9*1/*3 group. carboxy-celecoxib 12-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 26360837-11 2015 In terms of carboxy-celecoxib, the AUC0-48 was increased by 25.2%, the t1/2 prolonged 16.1% and the CL/F was decreased by 21.2% in CYP2C9*1/*3 group. Fluorine 103-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 26995878-4 2015 Recent pharmacogenomic (PGx) studies have shown that several single nucleotide polymorphisms (SNPs) in CYP2C9 (warfarin metabolic enzyme) and VKORC1 (warfarin target enzyme) are responsible for an individual"s warfarin sensitivity. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 26296708-1 2015 In this study, IC50 shift and time-dependent inhibition (TDI) experiments were carried out to measure the ability of amiodarone (AMIO), and its circulating human metabolites, to reversibly and irreversibly inhibit CYP1A2, CYP2C9, CYP2D6, and CYP3A4 activities in human liver microsomes. Amiodarone 117-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 26296708-1 2015 In this study, IC50 shift and time-dependent inhibition (TDI) experiments were carried out to measure the ability of amiodarone (AMIO), and its circulating human metabolites, to reversibly and irreversibly inhibit CYP1A2, CYP2C9, CYP2D6, and CYP3A4 activities in human liver microsomes. Amiodarone 129-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 222-228 26296708-3 2015 Based on these values, the minor metabolite N,N-didesethylamiodarone (DDEA) is predicted to be the major cause of DDIs with xenobiotics primarily metabolized by CYP1A2, CYP2C9, or CYP3A4, while AMIO and its N-monodesethylamiodarone (MDEA) derivative are the most likely cause of interactions involving inhibition of CYP2D6 metabolism. n,n-didesethylamiodarone 44-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 26296708-3 2015 Based on these values, the minor metabolite N,N-didesethylamiodarone (DDEA) is predicted to be the major cause of DDIs with xenobiotics primarily metabolized by CYP1A2, CYP2C9, or CYP3A4, while AMIO and its N-monodesethylamiodarone (MDEA) derivative are the most likely cause of interactions involving inhibition of CYP2D6 metabolism. Di-N-desethylamiodarone 70-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 26296708-3 2015 Based on these values, the minor metabolite N,N-didesethylamiodarone (DDEA) is predicted to be the major cause of DDIs with xenobiotics primarily metabolized by CYP1A2, CYP2C9, or CYP3A4, while AMIO and its N-monodesethylamiodarone (MDEA) derivative are the most likely cause of interactions involving inhibition of CYP2D6 metabolism. Amiodarone 194-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 26296708-3 2015 Based on these values, the minor metabolite N,N-didesethylamiodarone (DDEA) is predicted to be the major cause of DDIs with xenobiotics primarily metabolized by CYP1A2, CYP2C9, or CYP3A4, while AMIO and its N-monodesethylamiodarone (MDEA) derivative are the most likely cause of interactions involving inhibition of CYP2D6 metabolism. desethylamiodarone 207-231 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 26296708-3 2015 Based on these values, the minor metabolite N,N-didesethylamiodarone (DDEA) is predicted to be the major cause of DDIs with xenobiotics primarily metabolized by CYP1A2, CYP2C9, or CYP3A4, while AMIO and its N-monodesethylamiodarone (MDEA) derivative are the most likely cause of interactions involving inhibition of CYP2D6 metabolism. desethylamiodarone 233-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 169-175 26360116-7 2015 These findings provided for the development of a low-cost, simple, and fast direct whole blood genotyping method that uses Pfu DNA polymerase combined with phosphorothioate AS primers for CYP2C9*3 and VKORC1(-1639) loci. Parathion 156-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 26757860-2 2015 Pharmacogenetic studies have shown that common variants in genes CYP2C9 (*2 and *3) and VKORC1 (-1639G>A) are associated with lower requirements of OAC. SDZ 33-243 151-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 26433837-2 2015 Genetic polymorphisms in CYP2C9, VKORC1 and CYP4F2 have been shown to play an important role in variations in the warfarin maintenance dose in adults, but their effects in children remain unclear. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25929560-6 2015 Enzalutamide reduced the AUC of oral S-warfarin, omeprazole, and midazolam by 56, 70, and 86 %, respectively; therefore, enzalutamide is a moderate inducer of CYP2C9 and CYP2C19 and a strong inducer of CYP3A4. enzalutamide 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 26431339-9 2015 Sensitivity analysis suggested changes in CYP2C9 activity, and to a lesser extent UGT2B7, as the primary factor contributing to differences in indomethacin disposition between pregnancy and non-pregnancy. Indomethacin 143-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 26423799-5 2015 We found that the formation of 4OH-NDM-TOR was catalyzed both by CYP2C9 and CYP2D6, whereas the formation of 4OH-TAM and endoxifen was specifically catalyzed by CYP2D6 in HLMs. 4oh-ndm-tor 31-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 25929560-6 2015 Enzalutamide reduced the AUC of oral S-warfarin, omeprazole, and midazolam by 56, 70, and 86 %, respectively; therefore, enzalutamide is a moderate inducer of CYP2C9 and CYP2C19 and a strong inducer of CYP3A4. enzalutamide 122-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 160-166 25929560-8 2015 It is recommended to avoid concomitant use of enzalutamide with narrow therapeutic index drugs metabolized by CYP2C9, CYP2C19, or CYP3A4, as enzalutamide may decrease their exposure. enzalutamide 46-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 25929560-8 2015 It is recommended to avoid concomitant use of enzalutamide with narrow therapeutic index drugs metabolized by CYP2C9, CYP2C19, or CYP3A4, as enzalutamide may decrease their exposure. enzalutamide 141-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 26423799-5 2015 We found that the formation of 4OH-NDM-TOR was catalyzed both by CYP2C9 and CYP2D6, whereas the formation of 4OH-TAM and endoxifen was specifically catalyzed by CYP2D6 in HLMs. 4-hydroxy-N-desmethyltamoxifen 121-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 26233334-6 2015 Phenotyping of CYP2C9 was performed by administration of a 50-mg single oral dose of losartan and by calculating the urinary metabolic ratio (MR) of probe drug to its metabolite E-3174. Losartan 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 26418980-9 2015 VKORC1 -1639G/A, CYP2C9 430C/T, CYP2C9 1075A/C and CYP4F2 V433M polymorphisms have significant influence on stable warfarin dosage. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 26865929-0 2015 Identification of CYP2C9 and VKORC1 polymorphisms in Iranian patients who are under warfarin therapy. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 26865929-12 2015 CONCLUSION: In this study, samples were characterized by higher frequencies of CYP2C9*1 and VKORC1 G/A, determined as higher warfarin taking doses. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 26865929-13 2015 The results showed a significant relationship of the VCORC1 and CYP2C9 polymorphisms with warfarin sensitivity and severe side effects. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 26516586-1 2015 Our previous studies revealed that sesamin caused a mechanism-based inhibition (MBI) of CYP2C9 in human liver microsomes. sesamin 35-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 26516586-5 2015 These results suggest that the MI complex of cytochrome P450 and sesamin is unstable, and the effects of sesamin on human CYP2C9- or rat CYP2C-mediated drug metabolism may be small. sesamin 105-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 122-128 26837174-6 2015 Among them, the body weight (WT), CYP2C9 and VKORC1 genotype had conspicuous effect on warfarin PK/PD parameters. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 26418980-9 2015 VKORC1 -1639G/A, CYP2C9 430C/T, CYP2C9 1075A/C and CYP4F2 V433M polymorphisms have significant influence on stable warfarin dosage. Warfarin 115-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 24920353-1 2015 Rosuvastatin is used to treat dyslipidemia and is metabolized by CYP2C9 that shows variable metabolic activity in males and females. Rosuvastatin Calcium 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 26543813-0 2015 Adverse events in a newborn on valproate therapy due to loss-of-function mutations in CYP2C9. Valproic Acid 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 26543813-6 2015 The patient was demonstrated to carry two loss-of-function mutations in CYP2C9 (CYP2C9*3/*3) resulting in exaggerated blood concentrations of valproate. Valproic Acid 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 26543813-6 2015 The patient was demonstrated to carry two loss-of-function mutations in CYP2C9 (CYP2C9*3/*3) resulting in exaggerated blood concentrations of valproate. Valproic Acid 142-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 26543813-7 2015 The present case highlights the importance of assaying inborn errors in CYP2C9 gene in pediatric patients to avoid valproate-evoked serious side effects. Valproic Acid 115-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 26138612-8 2015 TMP 3"-demethylation was catalyzed by multiple P450s, including CYP2C9, correlated with CYP2C9 activity, and was inhibited by sulfaphenazole (up to 40%). Trimethoprim 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 26138612-8 2015 TMP 3"-demethylation was catalyzed by multiple P450s, including CYP2C9, correlated with CYP2C9 activity, and was inhibited by sulfaphenazole (up to 40%). Trimethoprim 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 26138612-8 2015 TMP 3"-demethylation was catalyzed by multiple P450s, including CYP2C9, correlated with CYP2C9 activity, and was inhibited by sulfaphenazole (up to 40%). Sulfaphenazole 126-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 26138612-9 2015 Overall, CYP2C9 and CYP3A4 appear to be the most significant contributors to TMP primary metabolism. Trimethoprim 77-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. Tamoxifen 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 26302287-3 2015 Warfarin metabolism is affected by variations in the cytochrome P450 2C9 (CYP2C9) and the vitamin K epoxide reductase complex 1 (VKORC1) genotypes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-72 26302287-3 2015 Warfarin metabolism is affected by variations in the cytochrome P450 2C9 (CYP2C9) and the vitamin K epoxide reductase complex 1 (VKORC1) genotypes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4'-hydroxytamoxifen 173-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 25994031-1 2015 CYP2C9, one of the most important drug-metabolizing enzymes, is responsible for metabolizing approximately 15% of clinically important drugs, including warfarin, diclofenac, and losartan. Warfarin 152-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25940823-3 2015 Tamoxifen is bioactivated by cytochrome P450 (CYP) enzymes such as CYP2B6, CYP2C9, CYP2C19, CYP2D6 and CYP3A4/5, resulting in the formation of active metabolites, including 4-hydroxy-tamoxifen and endoxifen. 4-hydroxy-N-desmethyltamoxifen 197-206 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 25994031-1 2015 CYP2C9, one of the most important drug-metabolizing enzymes, is responsible for metabolizing approximately 15% of clinically important drugs, including warfarin, diclofenac, and losartan. Diclofenac 162-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25994031-1 2015 CYP2C9, one of the most important drug-metabolizing enzymes, is responsible for metabolizing approximately 15% of clinically important drugs, including warfarin, diclofenac, and losartan. Losartan 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Isoleucine 35-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Isoleucine 35-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Isoleucine 35-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Phenylalanine 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Phenylalanine 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 162-168 25994031-4 2015 This base transversion leads to an Ile-to-Phe amino acid substitution at codon 434 within the CYP2C9 protein, and this new variant has been named a novel allele, CYP2C9*59, by the Human CYP Allele Nomenclature Committee (http://www.cypalleles.ki.se/cyp2c9.htm). Phenylalanine 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 249-255 25815675-0 2015 Improvements in CYP2C9 Genotyping Accuracy Are Needed: A Report of the First Proficiency Testing for Warfarin-related CYP2C9 and VKORC1 Genotyping in China. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25815675-0 2015 Improvements in CYP2C9 Genotyping Accuracy Are Needed: A Report of the First Proficiency Testing for Warfarin-related CYP2C9 and VKORC1 Genotyping in China. Warfarin 101-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25815675-2 2015 The cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) genotypes have been confirmed to be associated with warfarin dose requirements. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-23 25815675-2 2015 The cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex 1 (VKORC1) genotypes have been confirmed to be associated with warfarin dose requirements. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25904339-0 2015 Analysis of CYP2C9 polymorphisms (*2 and *3) in warfarin therapy patients in Pakistan. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25994870-0 2015 Association analysis of CYP2C9*3 and phenytoin-induced severe cutaneous adverse reactions (SCARs) in Thai epilepsy children. Phenytoin 37-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 25994870-6 2015 The association between CYP2C9*3 with phenytoin-induced SCARs and phenobarbital-induced SCARs were analyzed in comparison with tolerances and healthy samples. Phenytoin 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 25994870-7 2015 Significant association between phenytoin-induced SCARs and CYP2C9*3 was discovered (odds ratio=14.52; 95% confidence interval (CI)=1.18- , P-value=0.044). Phenytoin 32-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 25994870-9 2015 This study is the first report of CYP2C9*3 association to phenytoin-induced SCARs in Thai epileptic children. Phenytoin 58-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 25994870-10 2015 The CYP2C9*3 is a reasonable predictive genetic marker to anticipate SCARs from phenytoin. Phenytoin 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 26010205-0 2015 Influence of CYP2C9 polymorphism on the fall in International Normalized Ratio in patients interrupting warfarin therapy before elective surgery. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 26010205-3 2015 OBJECTIVES: The aim of this study was to investigate the potential influence of CYP2C9 polymorphism on the variable rate of fall in the International Normalized Ratio (INR) in patients withdrawing from warfarin treatment prior to elective surgery. Warfarin 202-210 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 26010205-9 2015 CONCLUSION: A genotype-guided protocol to tailor warfarin withdrawal according to an individual patient"s CYP2C9 genotype could reduce cancellation or delays of planned procedures, and could also be beneficial when transitioning patients from warfarin to one of the new oral anticoagulants. Warfarin 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 26010205-9 2015 CONCLUSION: A genotype-guided protocol to tailor warfarin withdrawal according to an individual patient"s CYP2C9 genotype could reduce cancellation or delays of planned procedures, and could also be beneficial when transitioning patients from warfarin to one of the new oral anticoagulants. Warfarin 243-251 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 25904339-1 2015 Association of CYP2C9 polymorphisms (*2 and*3) with warfarin dose, age, PT and INR. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 25904339-3 2015 Single nucleotide polymorphisms in hepatic VKORC1 and CYP2C9 genes causes decreased and increased metabolism of warfarin respectively. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 25904339-11 2015 Statistical analysis showed that low warfarin dose (weekly) is significantly associated with *1/*2 and *1/*3 genotypes (p value >= 0.001), whereas PT/INR showed no significant association with the any genotypes of CYP2C9. Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 25904339-12 2015 Our study suggest that polymorphic variants of CYP2C9 (*2 and *3) might influence warfarin dose requirements and associated with the low dose of warfarin in patients. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 25904339-12 2015 Our study suggest that polymorphic variants of CYP2C9 (*2 and *3) might influence warfarin dose requirements and associated with the low dose of warfarin in patients. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 26448196-2 2015 Genetic variants of the enzymes that metabolize warfarin, i.e. cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-82 26186657-0 2015 Clinical Practice Recommendations on Genetic Testing of CYP2C9 and VKORC1 Variants in Warfarin Therapy. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 26186657-4 2015 METHODS: A systematic literature search was performed for VKORC1 and CYP2C9 and their association with warfarin therapy. Warfarin 103-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 26448196-2 2015 Genetic variants of the enzymes that metabolize warfarin, i.e. cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 26448196-2 2015 Genetic variants of the enzymes that metabolize warfarin, i.e. cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses. Warfarin 194-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-82 26448196-2 2015 Genetic variants of the enzymes that metabolize warfarin, i.e. cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1), contribute to differences in patients" responses to various warfarin doses. Warfarin 194-202 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 25758405-3 2015 Both had variant cytochrome P2C9 (CYP2C9) alleles which reduce the metabolic capacity of the CYP2C9 enzyme responsible for the metabolism of the S-warfarin enantiomer. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-32 26024874-5 2015 Warfarin dose was influenced by age, BSA, CKD, amiodarone use, and CYP2C9*3 and VKORC1 variants in both races, by CYP2C9*2 and CYP4F2 variants in European Americans, and by rs12777823 in African Americans. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 26024874-5 2015 Warfarin dose was influenced by age, BSA, CKD, amiodarone use, and CYP2C9*3 and VKORC1 variants in both races, by CYP2C9*2 and CYP4F2 variants in European Americans, and by rs12777823 in African Americans. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 26469104-1 2015 BACKGROUND: Warfarin dose requirements are partly determined by common single nucleotide polymorphisms in VKORC1 and CYP2C9 genes. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 26469104-9 2015 CONCLUSIONS: The presence of CYP2C9*2 and/or *3 genotypes is associated with unstable warfarin treatment in patients after heart valve replacement, regardless of the type of INR testing. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 25758405-3 2015 Both had variant cytochrome P2C9 (CYP2C9) alleles which reduce the metabolic capacity of the CYP2C9 enzyme responsible for the metabolism of the S-warfarin enantiomer. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 25758405-3 2015 Both had variant cytochrome P2C9 (CYP2C9) alleles which reduce the metabolic capacity of the CYP2C9 enzyme responsible for the metabolism of the S-warfarin enantiomer. Warfarin 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 25758405-4 2015 Need for preoperative administration of vitamin K or postponement of an operation because of an INR >1.5 could be explained by variant alleles for CYP2C9 and age. Vitamin K 40-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 25808545-5 2015 We show that vitamin K2 and lithocholic acid, a by-product of intestinal flora, activate pregnane X receptor (PXR) and subsequent CYP3A4 and CYP2C9 expression in hPSC-derived and isolated fetal hepatocytes. Vitamin K 2 13-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 26125664-1 2015 Polymorphisms for VKORC1 and CYP2C9 are associated with increased warfarin sensitivity. Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 26125664-8 2015 One or more polymorphisms for VKORC1 or CYP2C9 (associated with warfarin sensitivity) were found in 67.7% of MCS patients. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 26125664-8 2015 One or more polymorphisms for VKORC1 or CYP2C9 (associated with warfarin sensitivity) were found in 67.7% of MCS patients. mcs 109-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 25699611-0 2015 Impact of CYP2C9 and VKORC1 genetic polymorphisms upon warfarin dose requirements in Egyptian patients with acute coronary syndrome. Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25699611-2 2015 Cytochrome P450-2C9 (CYP2C9) and vitamin K epoxide reductase-oxidaxe complex subunit 1 (VKORC1) contribute significantly to the variability of warfarin dose requirements among patients. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 25699611-2 2015 Cytochrome P450-2C9 (CYP2C9) and vitamin K epoxide reductase-oxidaxe complex subunit 1 (VKORC1) contribute significantly to the variability of warfarin dose requirements among patients. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 25699611-3 2015 We investigated the impact of CYP2C9 and VKORC1 polymorphisms on the variability of warfarin dosage requirements in Egyptian patients with acute coronary syndrome and their association with other nongenetic factors. Warfarin 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 25699611-9 2015 Regression analysis revealed that age, height, CYP2C9 and VKORC1 genotypes were significantly associated with warfarin dose. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 25699611-10 2015 Genetic polymorphisms in VKORC1, CYP2C9 along with age and height are determinants of warfarin dose requirements in Egyptian population acute coronary syndrome. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 25699611-11 2015 Higher warfarin loading dose is required for both wild CYP2C9 and VKORC1 gene variants which may contribute to warfarin-resistant cases. Warfarin 7-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 25699611-11 2015 Higher warfarin loading dose is required for both wild CYP2C9 and VKORC1 gene variants which may contribute to warfarin-resistant cases. Warfarin 111-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 25948712-8 2015 Although efavirenz is known as a marker substrate for human CYP2B6, efavirenz was not oxidized by CYP2B6 but by CYP2C9 in monkeys. efavirenz 68-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 25808545-5 2015 We show that vitamin K2 and lithocholic acid, a by-product of intestinal flora, activate pregnane X receptor (PXR) and subsequent CYP3A4 and CYP2C9 expression in hPSC-derived and isolated fetal hepatocytes. Lithocholic Acid 28-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 26142522-11 2015 Previous studies have also pointed out CYP2C9 polymorphism being responsible for such inter-individual differences in S-warfarin metabolism. Warfarin 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 25936586-3 2015 CTXA reversibly inhibited the CYP1A2-catalyzed phenacetin O-deethylation, CYP2C8-catalyzed paclitaxel 6-hydroxylation, and CYP2C9-catalyzed diclofenac 4"-hydroxylation with half-maximal inhibitory concentration (IC50) values of 3.9, 4.7, and 2.9 microM, respectively. cudratricusxanthone A 0-4 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 25943175-0 2015 Clinical relevance of genetic polymorphism in CYP2C9 gene to pharmacodynamics and pharmacokinetics of phenytoin in epileptic patients: validatory pharmacogenomic approach to pharmacovigilance. Phenytoin 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 25943175-10 2015 CYP2C9 genotype based phenytoin therapy is highly relevant in Kashmiri population due to a high incidence of genetic variations associated with therapeutic and adverse responses to phenytoin. Phenytoin 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25943175-10 2015 CYP2C9 genotype based phenytoin therapy is highly relevant in Kashmiri population due to a high incidence of genetic variations associated with therapeutic and adverse responses to phenytoin. Phenytoin 181-190 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26142522-12 2015 So plasma warfarin S/R ratio may serve as a useful phenotypic test for CYP2C9 polymorphism. Warfarin 10-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 25946405-0 2015 Variation in genes controlling warfarin disposition and response in American Indian and Alaska Native people: CYP2C9, VKORC1, CYP4F2, CYP4F11, GGCX. Warfarin 31-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 25946405-4 2015 Therefore, we were interested in variation in CYP2C9, VKORC1, CYP4F2, CYP4F11, and GGCX, which encode enzymes important for the activity of warfarin and synthesis of vitamin K-dependent blood clotting factors. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 25946405-4 2015 Therefore, we were interested in variation in CYP2C9, VKORC1, CYP4F2, CYP4F11, and GGCX, which encode enzymes important for the activity of warfarin and synthesis of vitamin K-dependent blood clotting factors. Vitamin K 166-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 26267945-4 2015 Cytochromes P450 (CYP) 2C9 and 3A4 are involved in the metabolism of tetrahydrocannabinol and cannabidiol, which implies possible DDI with CYP450 inhibitor and inducer, such as anticonvulsivants and HIV protease inhibitors, which may be prescribed in patients with neuropathic pain. Dronabinol 69-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-26 26267945-4 2015 Cytochromes P450 (CYP) 2C9 and 3A4 are involved in the metabolism of tetrahydrocannabinol and cannabidiol, which implies possible DDI with CYP450 inhibitor and inducer, such as anticonvulsivants and HIV protease inhibitors, which may be prescribed in patients with neuropathic pain. Cannabidiol 94-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-26 28930203-8 2015 Results: Vinpocetine showed a strong inhibition of P-gp (EC50 8 microM) and a moderate inhibition of recombinant CYP3A4 and CYP2D6 (IC50 2.8 and 6.5 microM) with no activity towards CYP2C9, CYP2C19 and CYP1A2 enzymes. vinpocetine 9-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 182-188 26179835-4 2015 The possible mechanism is speculated to be due to noni juiceinduced cytochrome P-450 2C9 metabolism of phenytoin. Phenytoin 103-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-88 25769357-13 2015 INTERPRETATION: CYP2C9 and VKORC1 genotypes identify patients who are more likely to experience early bleeding with warfarin and who derive a greater early safety benefit from edoxaban compared with warfarin. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25986145-2 2015 S-warfarin is metabolized to 7-S-hydroxywarfarin by the cytochrome P450 isoform 2C9 encoded by CYP2C9 gene. Warfarin 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 25712887-0 2015 Effects of CYP2C9*1/*3 genotype on the pharmacokinetics of flurbiprofen in Korean subjects. Flurbiprofen 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 25712887-1 2015 The aim of this study was to investigate the impact of CYP2C9*1/*3 genotype on the pharmacokinetics of flurbiprofen and its metabolite. Flurbiprofen 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 25712887-5 2015 AUCinf of flurbiprofen was significantly higher and its clearance was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. Flurbiprofen 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 25712887-5 2015 AUCinf of flurbiprofen was significantly higher and its clearance was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. Flurbiprofen 10-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 140-146 25712887-6 2015 The AUC ratio of 4"-hydroxyflurbiprofen to flurbiprofen was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. 4'-hydroxyflurbiprofen 17-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 25986145-2 2015 S-warfarin is metabolized to 7-S-hydroxywarfarin by the cytochrome P450 isoform 2C9 encoded by CYP2C9 gene. 7-s-hydroxywarfarin 29-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 25712887-6 2015 The AUC ratio of 4"-hydroxyflurbiprofen to flurbiprofen was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. 4'-hydroxyflurbiprofen 17-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 25712887-6 2015 The AUC ratio of 4"-hydroxyflurbiprofen to flurbiprofen was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. Flurbiprofen 27-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 25712887-6 2015 The AUC ratio of 4"-hydroxyflurbiprofen to flurbiprofen was significantly lower in the CYP2C9*1/*3 individuals than in those with CYP2C9*1/*1. Flurbiprofen 27-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 25986145-10 2015 The S-7-hydroxywarfarin was lower in carriers of the VKORC1 c.-1639A allele (p = 0.0005) and CYP2C9*3 (p = 0.047). 7-hydroxywarfarin 4-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 25712887-7 2015 These results indicate that the individuals carrying of CYP2C9*3 have significant reduction in flurbiprofen metabolism. Flurbiprofen 95-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 25986145-11 2015 The S-7-hydroxywarfarin/S-warfarin ratio was lower in the carriers of CYP2C9*3 (p = 0.008), but not in those with VKORC1 -c.1639A allele. 7-hydroxywarfarin 4-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Metronidazole 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-97 25986145-11 2015 The S-7-hydroxywarfarin/S-warfarin ratio was lower in the carriers of CYP2C9*3 (p = 0.008), but not in those with VKORC1 -c.1639A allele. Sulfur 4-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 25986145-11 2015 The S-7-hydroxywarfarin/S-warfarin ratio was lower in the carriers of CYP2C9*3 (p = 0.008), but not in those with VKORC1 -c.1639A allele. Warfarin 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Metronidazole 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-106 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Fluconazole 18-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-97 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Fluconazole 18-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-106 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-97 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Warfarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-106 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-97 26053779-4 2015 Metronidazole and fluconazole inhibit the metabolism of warfarin by blocking cytochrome P-450 2C9 (CYP-2C9), the major metabolic pathway of warfarin, with the end result being dramatic increases in patients" international normalized ratios (INRs) and potentially fatal bleeding. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-106 25967074-0 2015 Clinical significance of CYP2C9-status guided valproic acid therapy in children. Valproic Acid 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25967074-3 2015 Thus, patients" CYP2C9-status may account for the predisposition to adverse reactions, and testing CYP2C9-status may contribute to the improvement and rationalization of VPA therapy in children. Valproic Acid 170-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 25967074-6 2015 When the results of CYP2C9 genotyping and CYP2C9 expression were combined, the patients" VPA-metabolizing capacity was predicted, and VPA dosing was adjusted to the patients" CYP2C9-status. Valproic Acid 89-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 25967074-6 2015 When the results of CYP2C9 genotyping and CYP2C9 expression were combined, the patients" VPA-metabolizing capacity was predicted, and VPA dosing was adjusted to the patients" CYP2C9-status. Valproic Acid 89-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25967074-6 2015 When the results of CYP2C9 genotyping and CYP2C9 expression were combined, the patients" VPA-metabolizing capacity was predicted, and VPA dosing was adjusted to the patients" CYP2C9-status. Valproic Acid 89-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25967074-11 2015 SIGNIFICANCE: The knowledge of pediatric patients" CYP2C9-status can contribute to the optimization of VPA dosing and to the avoidance of misdosing-induced side effects. Valproic Acid 103-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 25884291-0 2015 In vitro assessment of 39 CYP2C9 variants found in the Chinese population on the metabolism of the model substrate fluoxetine and a summary of their effects on other substrates. Fluoxetine 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 26219158-1 2015 BACKGROUND: Polymorphisms in cytochrome P450 2C9 and vitamin K epoxide reductase complex, subunit 1 genes (CYP2C9 and VKORC1, respectively) were previously shown to affect the warfarin dose required in anticoagulant therapy of deep vein thrombosis (DVT). Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-48 26219158-1 2015 BACKGROUND: Polymorphisms in cytochrome P450 2C9 and vitamin K epoxide reductase complex, subunit 1 genes (CYP2C9 and VKORC1, respectively) were previously shown to affect the warfarin dose required in anticoagulant therapy of deep vein thrombosis (DVT). Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 26219158-3 2015 OBJECTIVE: To identify the effect of CYP2C9 and VKORC1 genetic variants on warfarin dosage in the Thai population with DVT. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 25884291-2 2015 The objective of this study was to assess the catalytic characteristics of 39 CYP2C9 isoforms found in the Chinese population and their effects on the metabolism of the model substrate fluoxetine in vitro. Fluoxetine 185-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 25884291-9 2015 In addition, 28 CYP2C9 isoforms including CYP2C9*3 exhibited a trend towards substrate inhibition for fluoxetine. Fluoxetine 102-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25884291-9 2015 In addition, 28 CYP2C9 isoforms including CYP2C9*3 exhibited a trend towards substrate inhibition for fluoxetine. Fluoxetine 102-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25884291-10 2015 WHAT IS NEW AND CONCLUSION: This study provides the most comprehensive data on the enzymatic activities associated with all reported CYP2C9 variants in the Chinese population with regard to the widely used antidepressant drug, fluoxetine. Fluoxetine 227-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 25884291-11 2015 Our data indicate that more attention should be paid to subjects carrying the corresponding infrequent CYP2C9 alleles when administering fluoxetine in the clinic. Fluoxetine 137-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 25858232-10 2015 Stratified analyses demonstrated increased alcohol-related major bleeding risk in patients on warfarin for >=1 year and in those with a low-dose genotype (CYP2C9*2/*3, VKORC1(1173G>A), CYP4F2*1), but not in other sub-groups evaluated. Alcohols 43-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 25977991-3 2015 CYP2C9 genotype-based dose recommendations (e.g., for warfarin) is advocated. Warfarin 54-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25977991-6 2015 CYP2C9 genotype and phenotype was determined in 148 Swedes and 146 Koreans using losartan as a probe. Losartan 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25977991-7 2015 CYP2C9 enzyme activity was assessed using urinary losartan/metabolite E-3174 ratio. Losartan 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25977991-7 2015 CYP2C9 enzyme activity was assessed using urinary losartan/metabolite E-3174 ratio. losartan carboxylic acid 70-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25077812-3 2015 CYP2C9 and CYP2E1 specifically metabolised sulfonylurea herbicides and halogenated hydrocarbons respectively. Sulfonylurea Compounds 43-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25077812-3 2015 CYP2C9 and CYP2E1 specifically metabolised sulfonylurea herbicides and halogenated hydrocarbons respectively. Hydrocarbons 83-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 29771647-0 2015 Phenoconversion of CYP2C9 in epilepsy limits the predictive value of CYP2C9 genotype in optimizing valproate therapy. Valproic Acid 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 29771647-0 2015 Phenoconversion of CYP2C9 in epilepsy limits the predictive value of CYP2C9 genotype in optimizing valproate therapy. Valproic Acid 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 29771647-1 2015 AIM: Since prominent role in valproate metabolism is assigned to CYP2C9 in pediatric patients, the association between children"s CYP2C9-status and serum valproate concentrations or dose-requirements was evaluated. Valproic Acid 29-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 29771647-1 2015 AIM: Since prominent role in valproate metabolism is assigned to CYP2C9 in pediatric patients, the association between children"s CYP2C9-status and serum valproate concentrations or dose-requirements was evaluated. Valproic Acid 154-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 29771647-2 2015 MATERIALS & METHODS: The contribution of CYP2C9 genotype and CYP2C9 expression in children (n = 50, Caucasian) with epilepsy to valproate pharmacokinetics was analyzed. Valproic Acid 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 29771647-3 2015 RESULTS: Valproate concentrations were significantly lower in normal expressers with CYP2C9*1/*1 than in low expressers or in patients carrying polymorphic CYP2C9 alleles. Valproic Acid 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 29771647-3 2015 RESULTS: Valproate concentrations were significantly lower in normal expressers with CYP2C9*1/*1 than in low expressers or in patients carrying polymorphic CYP2C9 alleles. Valproic Acid 9-18 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 29771647-6 2015 CONCLUSION: Due to the substantial downregulation of CYP2C9 expression in epilepsy, inferring patients" valproate metabolizing phenotype merely from CYP2C9 genotype results in false prediction. Valproic Acid 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 29771647-6 2015 CONCLUSION: Due to the substantial downregulation of CYP2C9 expression in epilepsy, inferring patients" valproate metabolizing phenotype merely from CYP2C9 genotype results in false prediction. Valproic Acid 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 29771648-2 2015 RESULTS & DISCUSSION: It was found that VKORC-1639G>A (25.5%), CYP2C9*2 (7.8%), CYP2C9*3 (6.1%), age (13.6%) and diagnosis (6.0%) significantly affected acenocoumarol dose variability in the derivation cohort. Adenosine Monophosphate 9-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 25747538-6 2015 The multiple linear regression model including VKORC1, CYP4F2, and CYP2C9 genetic variants, age, and weight could explain 42% of the variability in warfarin dosage. Warfarin 148-156 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 25499099-0 2015 Novel single nucleotide polymorphism in CYP2C9 is associated with changes in warfarin clearance and CYP2C9 expression levels in African Americans. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 25499099-1 2015 Warfarin is a widely used anticoagulant whose active S-enantiomer is primarily metabolized by the CYP2C9 enzyme. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 25499099-2 2015 The CYP2C9*2 and CYP2C9*3 alleles are associated with lower warfarin dose requirement and decreased enzyme activity. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 25499099-2 2015 The CYP2C9*2 and CYP2C9*3 alleles are associated with lower warfarin dose requirement and decreased enzyme activity. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 25499099-3 2015 In contrast, we previously identified a novel single-nucleotide polymorphism (SNP) (rs7089580A > T) in CYP2C9 that is associated with higher warfarin dose requirement in African Americans (AAs). Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 25499099-7 2015 Our findings indicate that rs7089580 is associated with higher S-warfarin clearance and CYP2C9 expression and may help explain the higher dose requirement of warfarin in AAs. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 25466603-1 2015 BACKGROUND AND OBJECTIVES: Approximately 50% of inter-individual variation in warfarin dose requirements is attributed to the polymorphisms of cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex, subunit 1 (VKORC1) genotypes. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-168 25466603-1 2015 BACKGROUND AND OBJECTIVES: Approximately 50% of inter-individual variation in warfarin dose requirements is attributed to the polymorphisms of cytochrome P450 (CYP) 2C9 (CYP2C9) and vitamin K epoxide reductase complex, subunit 1 (VKORC1) genotypes. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-176 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 25466603-7 2015 RESULTS: Using 437 warfarin measurements obtained from 325 patients, we found a correlation between plasma warfarin concentration and warfarin dose (r (2) = 0.356; P < 0.001) and a significant difference in the warfarin/7-hydroxywarfarin ratios of the CYP2C9*1/*1 and CYP2C9*1/*3 genotypes combined with drugs that inhibited warfarin (P = 0.003). Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 271-277 25585969-6 2015 RESULTS: Pharmacogenetic testing revealed a relation between this adverse event and an allelic variant of cytochrome P450, CYP2C9, subsequently leading to discontinuation of the drug along with counseling to caution the patient to avoid the use of celecoxib and other drugs metabolized by the same enzyme. Celecoxib 248-257 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 25515945-2 2015 The impact of activity of the genes encoding CYP3A4, CYP3A5, and CYP2C9 on the pharmacokinetics of voriconazole cannot be ignored because, second to CYP2C19, they are the most important enzymes involved in voriconazole metabolism. Voriconazole 99-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 25470432-8 2015 In both cells, the expressions of CYP2C8, CYP2C9, CYP3A4 and constitutive androstane receptor (CAR) were decreased by MTZ treatment. Metronidazole 118-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Metronidazole 50-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Metronidazole 50-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Warfarin 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Warfarin 58-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Metronidazole 87-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Metronidazole 87-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25470432-11 2015 Our findings suggest that the interaction between MTZ and S-warfarin may be due to the MTZ-induced down-regulation of CYP2C9, the primary enzyme responsible for S-warfarin hydroxylation, and CAR, which regulates CYP2C9 expression. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 212-218 25804257-6 2015 The inhibitor sulfaphenazole, in addition to the previously studied aniline, increased the barrier height for electron transfer and thereby makes CYP2C9 reduction more difficult and inhibits metabolism. Sulfaphenazole 14-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 25804257-6 2015 The inhibitor sulfaphenazole, in addition to the previously studied aniline, increased the barrier height for electron transfer and thereby makes CYP2C9 reduction more difficult and inhibits metabolism. aniline 68-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 25875957-0 2015 Correction: CYP2C9 genotype vs. metabolic phenotype for individual drug dosing--a correlation analysis using flurbiprofen as probe drug. Flurbiprofen 109-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25623616-9 2015 Compared to conventional in vitro BDI methodologies of assessment, the introduction of human plasma into the in vitro study model changed the observed inhibitory effect of silybin A, silybin B and hydrastine and berberine on CYP2C9 and CYP3A4/5, respectively, results which more closely mirrored those generated in clinical study. Silybin 172-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 225-231 25623616-9 2015 Compared to conventional in vitro BDI methodologies of assessment, the introduction of human plasma into the in vitro study model changed the observed inhibitory effect of silybin A, silybin B and hydrastine and berberine on CYP2C9 and CYP3A4/5, respectively, results which more closely mirrored those generated in clinical study. Silybin 183-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 225-231 25623616-9 2015 Compared to conventional in vitro BDI methodologies of assessment, the introduction of human plasma into the in vitro study model changed the observed inhibitory effect of silybin A, silybin B and hydrastine and berberine on CYP2C9 and CYP3A4/5, respectively, results which more closely mirrored those generated in clinical study. Berberine 212-221 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 225-231 24517573-0 2015 Inhibitory effects of curcumin on activity of cytochrome P450 2C9 enzyme in human and 2C11 in rat liver microsomes. Curcumin 22-30 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-65 24517573-1 2015 Cytochrome P450 2C9 (CYP2C9), one of the most important phase I drug metabolizing enzymes, could catalyze the reactions that convert diclofenanc into diclofenac 4"-hydroxylation. diclofenanc 133-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24517573-1 2015 Cytochrome P450 2C9 (CYP2C9), one of the most important phase I drug metabolizing enzymes, could catalyze the reactions that convert diclofenanc into diclofenac 4"-hydroxylation. diclofenanc 133-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24517573-1 2015 Cytochrome P450 2C9 (CYP2C9), one of the most important phase I drug metabolizing enzymes, could catalyze the reactions that convert diclofenanc into diclofenac 4"-hydroxylation. diclofenac 4" 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24517573-1 2015 Cytochrome P450 2C9 (CYP2C9), one of the most important phase I drug metabolizing enzymes, could catalyze the reactions that convert diclofenanc into diclofenac 4"-hydroxylation. diclofenac 4" 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24517573-3 2015 The objective of this work was to investigate the effects of curcumin on CYP2C9 in human and cytochrome P450 2C11 (CYP2C11) in rat liver microsomes. Curcumin 61-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 24517573-4 2015 The results showed that curcumin inhibited CYP2C9 activity (10 micromol L(-1) diclofenac) with half-maximal inhibition or a half-maximal inhibitory concentration (IC50) of 15.25 micromol L(-1) and Ki = 4.473 micromol L(-1) in human liver microsomes. Curcumin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 24517573-4 2015 The results showed that curcumin inhibited CYP2C9 activity (10 micromol L(-1) diclofenac) with half-maximal inhibition or a half-maximal inhibitory concentration (IC50) of 15.25 micromol L(-1) and Ki = 4.473 micromol L(-1) in human liver microsomes. Diclofenac 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 24517573-5 2015 Curcumin"s mode of action on CYP2C9 activity was noncompetitive for the substrate diclofenanc and uncompetitive for the cofactor NADPH. Curcumin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 24517573-5 2015 Curcumin"s mode of action on CYP2C9 activity was noncompetitive for the substrate diclofenanc and uncompetitive for the cofactor NADPH. diclofenanc 82-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 24517573-5 2015 Curcumin"s mode of action on CYP2C9 activity was noncompetitive for the substrate diclofenanc and uncompetitive for the cofactor NADPH. NADP 129-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 24517573-7 2015 The observations imply that curcumin has the inhibitory effects on CYP2C9 activity in human. Curcumin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 25652102-6 2015 The genotypes of VKORC1, CYP2C9, and CYP2C19, together with non-genetic factors (age, sex, and body weight) explained 52% of the variability in warfarin INR/daily dose, of which CYP2C19 genotypes accounted for 7%. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25515945-2 2015 The impact of activity of the genes encoding CYP3A4, CYP3A5, and CYP2C9 on the pharmacokinetics of voriconazole cannot be ignored because, second to CYP2C19, they are the most important enzymes involved in voriconazole metabolism. Voriconazole 206-218 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 25515945-3 2015 The influence of genetic polymorphisms in CYP3A4, CYP3A5, and CYP2C9 on the plasma concentrations of voriconazole was evaluated in the present study. Voriconazole 101-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 25656643-6 2015 The specific cytochrome P450 (CYP) 2C9 inhibitor, sulfaphenazole, counteracted both oxidative stress and mitochondrial membrane depolarization, providing EC protection against resveratrol-elicited pro-oxidant effects. Sulfaphenazole 50-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-38 25656643-6 2015 The specific cytochrome P450 (CYP) 2C9 inhibitor, sulfaphenazole, counteracted both oxidative stress and mitochondrial membrane depolarization, providing EC protection against resveratrol-elicited pro-oxidant effects. Resveratrol 176-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-38 25656643-7 2015 Our findings strongly suggest that CYP2C9 mediates resveratrol-induced oxidative stress leading to mitochondria impairment and EC death. Resveratrol 51-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 25449227-2 2015 In vitro studies indicated that faldaprevir inhibited CYP2B6, CYP2C9, and CYP3A, and was a weak-to-moderate inactivator of CYP3A4. faldaprevir 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 25595525-1 2015 INTRODUCTION: A large proportion of the coumarin dose variability is explained by environmental factors and by common genetic variants in the VKORC1 and CYP2C9 genes. coumarin 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 153-159 25449227-6 2015 These studies suggest that faldaprevir shows a dose-dependent inhibition of CYP3A and CYP2C9, and does not induce CYP isoforms. faldaprevir 27-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 28294900-2 2015 In the group with PG selection we took into consideration the result of preliminary genotyping of polymorphisms of VKORC1 and CYP2C9 genes known to be associated with individual warfarin dose. Warfarin 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 28294900-6 2015 Carriers of *2 and/or *3 of CYP2C9 associated with lowering of activity of this cytochrome had greater lability of INR values during course of therapy with warfarin. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 25449654-1 2015 The induction of CYP2C9 by dabrafenib using S-warfarin as a probe and the effects of a CYP3A inhibitor (ketoconazole) and a CYP2C8 inhibitor (gemfibrozil) on dabrafenib pharmacokinetics were evaluated in patients with BRAF V600 mutation-positive tumors. dabrafenib 27-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 25162219-0 2015 Individualized phenytoin therapy for Japanese pediatric patients with epilepsy based on CYP2C9 and CYP2C19 genotypes. Phenytoin 15-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 25162219-1 2015 BACKGROUND: The aims of this study were to identify the target dose of phenytoin (PHT) and to compare the treatment continuation rate between patients receiving conventional therapy and patients receiving individualized therapy based on genotyping of the CYP2C9*3, CYP2C19*2, and CYP2C19*3 alleles. Phenytoin 71-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 25162219-6 2015 If the patients had the CYP2C9*3, CYP2C19*2, or CYP2C19*3 alleles, the initial dose of PHT was reduced by 10%-50% according to previous reports. Phenytoin 87-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 25775139-0 2015 CYP2C9 genotype vs. metabolic phenotype for individual drug dosing--a correlation analysis using flurbiprofen as probe drug. Flurbiprofen 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25758259-4 2015 Owing to the unique hydrophobic feature of a ZnO NW that provides a desirable "microenvironment" for the immobilization of biomolecules, our device can effectively stimulate the tolbutamide metabolism by decorating a ZnO NW with cytochrome P4502C9/CYPs reductase (CYP2C9/CPR) microsomes. Zinc Oxide 45-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 264-270 25758259-4 2015 Owing to the unique hydrophobic feature of a ZnO NW that provides a desirable "microenvironment" for the immobilization of biomolecules, our device can effectively stimulate the tolbutamide metabolism by decorating a ZnO NW with cytochrome P4502C9/CYPs reductase (CYP2C9/CPR) microsomes. Tolbutamide 178-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 264-270 25758259-4 2015 Owing to the unique hydrophobic feature of a ZnO NW that provides a desirable "microenvironment" for the immobilization of biomolecules, our device can effectively stimulate the tolbutamide metabolism by decorating a ZnO NW with cytochrome P4502C9/CYPs reductase (CYP2C9/CPR) microsomes. Zinc Oxide 217-220 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 264-270 25560189-0 2015 Genotype frequencies of VKORC1 and CYP2C9 in native and Mestizo populations from Mexico, potential impact for coumarin dosing. coumarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-41 25304014-0 2015 Effect of CYP2C9, CYP4F2 and VKORC1 genetic polymorphisms on pharmacokinetics and pharmacodynamics of mean daily maintenance dose of warfarin in Chinese patients. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25578229-9 2015 In PHH, a low CYP2C9 activity, the major IBU-metabolising CYP, led to an increased cytotoxicity. Ibuprofen 41-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 25304014-7 2015 There were significant differences in the concentrations of S-WF and 7-OH-S-WF among the CYP2C9 variants. 7-oh-s 69-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 25669612-1 2015 OBJECTIVE: Our aim was to determinate the acenocoumarol dose requirement in highly sensitive geriatric patients, based on a minimum of genotype (VKORC1 and CYP2C9) data. Acenocoumarol 42-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 25618289-6 2015 Genetic variants of other CYP enzymes, including CYP3A4 and CYP2C9/19, as well as co-medication interfering with the metabolic activity of CYP2D6 and CYP3A4 have been shown to affect endoxifen concentrations and may also contribute to the variability in response to tamoxifen. 4-hydroxy-N-desmethyltamoxifen 183-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 25618289-6 2015 Genetic variants of other CYP enzymes, including CYP3A4 and CYP2C9/19, as well as co-medication interfering with the metabolic activity of CYP2D6 and CYP3A4 have been shown to affect endoxifen concentrations and may also contribute to the variability in response to tamoxifen. Tamoxifen 266-275 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 25187307-0 2015 Effect of CYP2C9-VKORC1 interaction on warfarin stable dosage and its predictive algorithm. Warfarin 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25187307-1 2015 This study aimed to identify the effect of CYP2C9-VKORC1 interaction on warfarin dosage requirement and its predictive algorithm by investigating four populations. Warfarin 72-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 25311916-8 2015 The highest mean phenytoin level (27.95 +- 1.85 microg/ml) was detected in the G8 genotype group (CYP2C9*1/*3-CYP2C19*2/*3) and the G1 genotype group showed the lowest mean phenytoin level (7.43 +- 0.73 microg/ml). Phenytoin 17-26 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 98-104 28962416-0 2015 In vitro modulatory effects of Terminalia arjuna, arjunic acid, arjunetin and arjungenin on CYP3A4, CYP2D6 and CYP2C9 enzyme activity in human liver microsomes. arjunic acid 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 28962416-0 2015 In vitro modulatory effects of Terminalia arjuna, arjunic acid, arjunetin and arjungenin on CYP3A4, CYP2D6 and CYP2C9 enzyme activity in human liver microsomes. arjunetin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 28962416-3 2015 Alcoholic and aqueous bark extracts of T. arjuna, arjunic acid, arjunetin and arjungenin were evaluated for their potential to inhibit CYP3A4, CYP2D6 and CYP2C9 enzymes in human liver microsomes. arjunic acid 50-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 28962416-3 2015 Alcoholic and aqueous bark extracts of T. arjuna, arjunic acid, arjunetin and arjungenin were evaluated for their potential to inhibit CYP3A4, CYP2D6 and CYP2C9 enzymes in human liver microsomes. arjunetin 64-73 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 28962416-3 2015 Alcoholic and aqueous bark extracts of T. arjuna, arjunic acid, arjunetin and arjungenin were evaluated for their potential to inhibit CYP3A4, CYP2D6 and CYP2C9 enzymes in human liver microsomes. arjungenin 78-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 28962416-7 2015 Our findings suggest strongly that arjuna extracts significantly inhibit the activity of CYP3A4, CYP2D6 and CYP2C9 enzymes, which is likely to cause clinically significant drug-drug interactions mediated via inhibition of the major CYP isozymes. arjuna 35-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 25663116-2 2015 The dosage of warfarin is strongly affected by genetic variants of CYP2C9 and VKORC1 genes. Warfarin 14-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 25597548-3 2015 Genetic polymorphisms of CYP2C9 and 2C19 are reasonably common and further prolong the elimination of phenytoin. Phenytoin 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-40 25533629-7 2015 VMY-2-95 2HCl showed significant inhibition of CYP2C9 and 2C19, but weak or no effect on 1A2, 2D6, and 3A4. vmy-2-95 2hcl 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 25052959-3 2015 HS-23 slightly inhibited CYP2A6, CYP2B6, CYP2C9, CYP2C19, and CYP3A4 enzyme activities in human liver microsomes with IC50 values of 80.6, 160.7, 169.5, 85.4, and 76.6 mug/mL, respectively. hs-23 0-5 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 25681367-7 2015 Phenytoin is metabolized in the liver, primarily by the CYP2C9 isozyme, which can be competitively inhibited by capecitabine. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 25681367-7 2015 Phenytoin is metabolized in the liver, primarily by the CYP2C9 isozyme, which can be competitively inhibited by capecitabine. Capecitabine 112-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Simvastatin 47-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25181036-8 2015 Genetic polymorphisms in the gene encoding cytochrome P450 2C9 (CYP2C9) and those in the target gene responsible for the warfarin anticoagulant effect, vitamin K epoxide reductase (VKORC1), account for much of the variability in the warfarin maintenance dose; however, routine genotyping in warfarin therapy remains controversial. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 25181036-8 2015 Genetic polymorphisms in the gene encoding cytochrome P450 2C9 (CYP2C9) and those in the target gene responsible for the warfarin anticoagulant effect, vitamin K epoxide reductase (VKORC1), account for much of the variability in the warfarin maintenance dose; however, routine genotyping in warfarin therapy remains controversial. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 25181036-8 2015 Genetic polymorphisms in the gene encoding cytochrome P450 2C9 (CYP2C9) and those in the target gene responsible for the warfarin anticoagulant effect, vitamin K epoxide reductase (VKORC1), account for much of the variability in the warfarin maintenance dose; however, routine genotyping in warfarin therapy remains controversial. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-62 25181036-8 2015 Genetic polymorphisms in the gene encoding cytochrome P450 2C9 (CYP2C9) and those in the target gene responsible for the warfarin anticoagulant effect, vitamin K epoxide reductase (VKORC1), account for much of the variability in the warfarin maintenance dose; however, routine genotyping in warfarin therapy remains controversial. Warfarin 233-241 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Amiodarone 106-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. MDEA 115-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Dextromethorphan 148-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Amiodarone 207-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. MDEA 268-272 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Warfarin 308-316 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. Amiodarone 207-211 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25324279-7 2015 The closest prediction was obtained for CYP3A (simvastatin) DDI when the competitive inhibition from both AMIO and MDEA was considered, for CYP2D6 (dextromethorphan) DDI when the competitive inhibition from AMIO and the competitive plus time-dependent inhibition from MDEA were incorporated, and for CYP2C9 (warfarin) DDI when the competitive plus time-dependent inhibition from AMIO and the competitive inhibition from MDEA were considered. MDEA 268-272 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 300-306 25760542-2 2015 Cynomolgus CYP2C9 (formerly known as CYP2C43) is predominantly expressed in liver and encodes a drug-metabolizing enzyme that metabolizes human CYP2C substrates such as S-mephenytoin and progesterone. Mephenytoin 169-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 25480923-7 2015 Enzyme activities of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were reduced upon 48-72 hours exposure to IL-6 in PHH and HepaRG. 4,5,6,7-tetrachlorophthalide 123-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 25760542-2 2015 Cynomolgus CYP2C9 (formerly known as CYP2C43) is predominantly expressed in liver and encodes a drug-metabolizing enzyme that metabolizes human CYP2C substrates such as S-mephenytoin and progesterone. Mephenytoin 169-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-16 25480923-7 2015 Enzyme activities of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, and CYP3A4 were reduced upon 48-72 hours exposure to IL-6 in PHH and HepaRG. heparg 131-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 25760542-2 2015 Cynomolgus CYP2C9 (formerly known as CYP2C43) is predominantly expressed in liver and encodes a drug-metabolizing enzyme that metabolizes human CYP2C substrates such as S-mephenytoin and progesterone. Progesterone 187-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 25760542-2 2015 Cynomolgus CYP2C9 (formerly known as CYP2C43) is predominantly expressed in liver and encodes a drug-metabolizing enzyme that metabolizes human CYP2C substrates such as S-mephenytoin and progesterone. Progesterone 187-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-16 25760542-3 2015 In addition, cynomolgus CYP2C9 also metabolizes caffeine, resulting in the formation of the metabolite that is not generated efficiently in humans. Caffeine 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24962733-1 2015 Previous studies have raised interest in using the genotyping of CYP2C9 and VKORC1 to guide warfarin dosing. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 25519826-1 2015 OBJECTIVE: The objective of this study was to determine the influence of CYP2C9, VKORC1, CYP4F2, and GGCX genetic polymorphisms on mean daily dose of acenocoumarol in South Indian patients and to develop a new pharmacogenetic algorithm based on clinical and genetic factors. Acenocoumarol 150-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 25519826-6 2015 The genetic variants of CYP2C9 and GGCX (rs11676382) were found to be associated with lower acenocoumarol dose, whereas CYP4F2 (rs2108622) was associated with higher doses. Acenocoumarol 92-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 25519826-7 2015 Age, body mass index (BMI), variation of CYP2C9, VKORC1, CYP4F2, and GGCX were the major determinants of acenocoumarol maintenance dose, accounting for 61.8 % of its variability (adjusted r (2) = 0.615, p < 0.0001). Acenocoumarol 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 25727956-7 2015 CYP2C9 and CYP2C19 activities were inhibited down to 75-80% (at the same rocuronium concentration). Rocuronium 73-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25026456-1 2015 Interindividual variability in stable warfarin doses is largely attributed to VKORC1 and CYP2C9 variants. Warfarin 38-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 25026456-5 2015 This study revealed that GATA4 can be predictive of stable warfarin dose and extended warfarin pharmacogenetics further to the regulation of CYP2C9 expression. Warfarin 59-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 25026456-5 2015 This study revealed that GATA4 can be predictive of stable warfarin dose and extended warfarin pharmacogenetics further to the regulation of CYP2C9 expression. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 141-147 25091503-4 2015 CYP2C19 and CYP2C9 correlated with norendoxifen and (Z)-4-hydroxytamoxifen concentrations, respectively (P<0.001). N,N-didesmethyl-4-hydroxytamoxifen 35-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25560582-3 2015 CYP2C8, CYP2C9 and CYP2J2 gene involved in the metabolism of arachidonic acid, generates epoxyeicosatrienoic acids (EETs) that mediate dilation of coronary arteries improving post-ischemic cardiac contractile function, reduce vascular inflammation, and increase intravascular fibrinolysis. Arachidonic Acid 61-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 25560582-3 2015 CYP2C8, CYP2C9 and CYP2J2 gene involved in the metabolism of arachidonic acid, generates epoxyeicosatrienoic acids (EETs) that mediate dilation of coronary arteries improving post-ischemic cardiac contractile function, reduce vascular inflammation, and increase intravascular fibrinolysis. epoxyeicosatrienoic acids 89-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 25091503-4 2015 CYP2C19 and CYP2C9 correlated with norendoxifen and (Z)-4-hydroxytamoxifen concentrations, respectively (P<0.001). hydroxytamoxifen 52-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25462116-7 2015 CYP2C9 and CYP3A4, which metabolize DF to p-benzoquinone imines, were tested to investigate the applicability of human P450s. 1,4-benzoquinoneimine 42-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26632190-1 2015 Arachidonic acid (AA) is metabolized to epoxyeicosatrienoic acids (EETs) via cytochrome enzymes such as CYP 2C9, 2C8 and 2J2. Arachidonic Acid 0-16 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-111 25391650-0 2015 Epigenetic modification of histone 3 lysine 27: mediator subunit MED25 is required for the dissociation of polycomb repressive complex 2 from the promoter of cytochrome P450 2C9. Lysine 37-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-177 25391650-9 2015 Formaldehyde-assisted isolation of regulatory elements (FAIRE) revealed chromatin conformation changes that were reliant on MED25, indicating that MED25 induced a permissive chromatin state that reflected increases in CYP2C9 mRNA. Formaldehyde 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 26632190-1 2015 Arachidonic acid (AA) is metabolized to epoxyeicosatrienoic acids (EETs) via cytochrome enzymes such as CYP 2C9, 2C8 and 2J2. epoxyeicosatrienoic acids 40-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-111 26632190-1 2015 Arachidonic acid (AA) is metabolized to epoxyeicosatrienoic acids (EETs) via cytochrome enzymes such as CYP 2C9, 2C8 and 2J2. eets 67-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-111 25832633-0 2015 Impact of CYP2C9 polymorphism found in the Chinese population on the metabolism of propofol in vitro. Propofol 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25832633-1 2015 The microsomal CYP2C9 alleles involved in the biotransformation of propofol, a widely used anesthetic agent, were investigated in vitro. Propofol 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 25832633-2 2015 To examine the enzymatic activity of the CYP2C9 alleles, kinetic parameters for propofol 4-hydroxylation were determined in recombinant human P450s CYP2C9 microsomes from Sf21 insects cells carrying CYP2C9*1 and other variants. Propofol 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 25832633-6 2015 This study is the first to report these rare alleles for propofol metabolism, providing fundamental data for further clinical studies on CYP2C9 alleles for propofol metabolism in vivo. Propofol 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 25832633-6 2015 This study is the first to report these rare alleles for propofol metabolism, providing fundamental data for further clinical studies on CYP2C9 alleles for propofol metabolism in vivo. Propofol 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 137-143 26554252-0 2015 CYP2C9 Genetic Polymorphism is a Potential Predictive Marker for the Efficacy of Rosuvastatin Therapy. Rosuvastatin Calcium 81-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25757926-0 2015 In vitro inhibition of CYP2C9-mediated warfarin 7-hydroxylation by iguratimod: possible mechanism of iguratimod-warfarin interaction. Warfarin 39-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 25757926-0 2015 In vitro inhibition of CYP2C9-mediated warfarin 7-hydroxylation by iguratimod: possible mechanism of iguratimod-warfarin interaction. Warfarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 25757926-13 2015 These results indicated that iguratimod is a potent direct inhibitor of CYP2C9-mediated warfarin 7-hydroxylation and that its inhibitory effect on CYP2C9.1 was more sensitive than that on CYP2C9.3. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 26554252-3 2015 This study aimed to evaluate the associations of CYP2C9 genetic polymorphism with the efficacy and safety of rosuvastatin in Chinese patients with hyperlipidemia. Rosuvastatin Calcium 109-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 26554252-15 2015 CONCLUSIONS: This study suggests that the CYP2C9 polymorphism may be involved in the lipid-lowering efficacy of rosuvastatin in patients with hyperlipidemia. Rosuvastatin Calcium 112-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25144335-0 2015 Effect of CYP2C9 genetic polymorphism on the metabolism of flurbiprofen in vitro. Flurbiprofen 59-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25144335-2 2015 To examine the enzymatic activity of the CYP2C9 alleles, kinetic parameters for 4"-hydroxyflurbiprofen were determined using recombinant human P450s CYP2C9 microsomes from insect cells Sf21 carrying wild-type CYP2C9*1 and other variants. 4'-hydroxyflurbiprofen 80-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 25144335-2 2015 To examine the enzymatic activity of the CYP2C9 alleles, kinetic parameters for 4"-hydroxyflurbiprofen were determined using recombinant human P450s CYP2C9 microsomes from insect cells Sf21 carrying wild-type CYP2C9*1 and other variants. 4'-hydroxyflurbiprofen 80-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 25144335-2 2015 To examine the enzymatic activity of the CYP2C9 alleles, kinetic parameters for 4"-hydroxyflurbiprofen were determined using recombinant human P450s CYP2C9 microsomes from insect cells Sf21 carrying wild-type CYP2C9*1 and other variants. 4'-hydroxyflurbiprofen 80-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 25144335-5 2015 This study provided the most comprehensive data on the enzymatic activities of all reported CYP2C9 variants in the Chinese population with regard to the commonly used non-steroidal anti-inflammatory drug, flurbiprofen (FP). Flurbiprofen 205-217 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 26037375-2 2015 This article reviews the contribution of genetic polymorphisms affecting the activity of CYP2C9, the main enzyme responsible for phenytoin metabolism, to the variation in phenytoin clearance and susceptibility to adverse effects. Phenytoin 129-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 26037375-2 2015 This article reviews the contribution of genetic polymorphisms affecting the activity of CYP2C9, the main enzyme responsible for phenytoin metabolism, to the variation in phenytoin clearance and susceptibility to adverse effects. Phenytoin 171-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 26037375-3 2015 AREAS COVERED: Comprehensive and critical review of available evidence concerning the influence of CYP2C9 genetic polymorphism on phenytoin pharmacokinetic and safety profile. Phenytoin 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 26037375-4 2015 EXPERT OPINION: There is extensive evidence that CYP2C9 polymorphisms are an important determinant of the rate of phenytoin metabolism, although other factors including expression of other enzymes such as CYP2C19 and the influence of drug interactions, physiological and disease-related factors may also play a role. Phenytoin 114-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 26037375-6 2015 The clinical value and cost-effectiveness of CYP2C9 genotyping in improving the safety of phenytoin therapy, however, have not been clearly established and require formal testing in well-designed prospective studies. Phenytoin 90-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 26058399-6 2015 Fluvastatin is metabolized by CYP2C9, while pravastatin, rosuvastatin and pitavastatin are not susceptible to inhibition by any CYP. Fluvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 26502504-2 2015 In the group with PG selection we took into consideration the result of preliminary genotyping of polymorphisms of VKORC1 and CYP2C9 genes known to be associated with individual warfarin dose. Warfarin 178-186 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 25345887-4 2015 EXPERT OPINION: Gene candidate studies in Brazilian patients verified consistently the association of warfarin and pheprocoumon stable dose requirements with CYP2C9 and VKORC1 polymorphisms, and minor or no influence of other pharmacogenes (e.g., CYP4F2 and F7). Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 25103957-2 2015 A series of in vitro experiments conducted early in the development of ivacaftor indicated ivacaftor and metabolites may have the potential to inhibit cytochrome P450 (CYP) 2C8, CYP2C9, CYP3A, and CYP2D6, as well as P-glycoprotein (P-gp). ivacaftor 91-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 178-184 25103957-7 2015 Based on these results, caution and appropriate monitoring are recommended when concomitant substrates of CYP2C9, CYP3A and/or P-gp are used during treatment with ivacaftor, particularly drugs with a narrow therapeutic index, such as warfarin. ivacaftor 163-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 25103957-7 2015 Based on these results, caution and appropriate monitoring are recommended when concomitant substrates of CYP2C9, CYP3A and/or P-gp are used during treatment with ivacaftor, particularly drugs with a narrow therapeutic index, such as warfarin. Warfarin 234-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 106-112 26517138-0 2015 The Implication of the Polymorphisms of COX-1, UGT1A6, and CYP2C9 among Cardiovascular Disease (CVD) Patients Treated with Aspirin. Aspirin 123-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 26517138-1 2015 PURPOSE: Enzymes potentially responsible for the pharmacokinetic variations of aspirin include cyclooxygenase-1 (COX-1), UDP-glucuronosyltransferase (UGT1A6) and P450 (CYP) (CYP2C9). Aspirin 81-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 26517138-10 2015 CONCLUSION: Screening of patients with defective genetic variants of UGT1A6 and CYP2C9*3 helps in identifying patients at risk of aspirin induced gastritis. Aspirin 130-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 26502504-6 2015 Carriers of *2 and/or *3 of CYP2C9 associated with lowering of activity of this cytochrome had greater lability of INR values during course of therapy with warfarin. Warfarin 156-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 25823787-0 2015 Mechanistic insights into the effect of CYP2C9*2 and CYP2C9*3 variants on the 7-hydroxylation of warfarin. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 25560470-5 2015 Polymorphisms of genes involved in drug metabolism, such as CYP2C9, CYP2C8 and SLCO1B1, may influence the efficacy of glinides and the incidence of adverse effects. glinides 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 25823787-0 2015 Mechanistic insights into the effect of CYP2C9*2 and CYP2C9*3 variants on the 7-hydroxylation of warfarin. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 25823787-1 2015 AIM: To evaluate the impact of CYP2C9*2 and CYP2C9*3 variants on binding and hydroxylation of warfarin. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 25823787-1 2015 AIM: To evaluate the impact of CYP2C9*2 and CYP2C9*3 variants on binding and hydroxylation of warfarin. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 55-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). 7-hydroxywarfarin 64-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 26122864-0 2015 Effect of gender and CYP2C9 and CYP2C8 polymorphisms on the pharmacokinetics of ibuprofen enantiomers. Ibuprofen 80-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). 7-hydroxywarfarin 64-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 26122864-1 2015 AIM: To evaluate the effect of polymorphisms in CYP2C9 and CYP2C8 and gender on the pharmacokinetics of the enantiomeric forms of ibuprofen. Ibuprofen 130-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 26122864-4 2015 CYP2C9*3 and CYP2C9*2 carriers had a lower S-ibuprofen clearance and a higher S-ibuprofen AUC and half-life. Ibuprofen 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). 7-hydroxywarfarin 64-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 26122864-4 2015 CYP2C9*3 and CYP2C9*2 carriers had a lower S-ibuprofen clearance and a higher S-ibuprofen AUC and half-life. Ibuprofen 43-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Hydrogen 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 26122864-4 2015 CYP2C9*3 and CYP2C9*2 carriers had a lower S-ibuprofen clearance and a higher S-ibuprofen AUC and half-life. Ibuprofen 78-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 26122864-4 2015 CYP2C9*3 and CYP2C9*2 carriers had a lower S-ibuprofen clearance and a higher S-ibuprofen AUC and half-life. Ibuprofen 78-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Hydrogen 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 26122864-5 2015 R-ibuprofen clearance was decreased in CYP2C9*3 carriers. Ibuprofen 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 26122864-7 2015 Multiple regression analysis showed that CYP2C9*2, CYP2C9*3 and gender were associated with S-ibuprofen clearance, but only CYP2C9*3 was associated with R-ibuprofen clearance. Ibuprofen 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Hydrogen 147-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 26122864-7 2015 Multiple regression analysis showed that CYP2C9*2, CYP2C9*3 and gender were associated with S-ibuprofen clearance, but only CYP2C9*3 was associated with R-ibuprofen clearance. Ibuprofen 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 26122864-7 2015 Multiple regression analysis showed that CYP2C9*2, CYP2C9*3 and gender were associated with S-ibuprofen clearance, but only CYP2C9*3 was associated with R-ibuprofen clearance. Ibuprofen 92-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 26122864-8 2015 CONCLUSION: The pharmacokinetics of S-ibuprofen and R-ibuprofen is affected by CYP2C9 polymorphisms and gender. Ibuprofen 36-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 26122864-8 2015 CONCLUSION: The pharmacokinetics of S-ibuprofen and R-ibuprofen is affected by CYP2C9 polymorphisms and gender. Ibuprofen 52-63 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-4 2015 RESULTS: CYP2C9*2 and CYP2C9*3 variants exhibited high warfarin/7-hydroxywarfarin (multiple linear regression model), dose-dependent disruption of hydrogen bonds with warfarin, dose-dependent increase in the distance between C7 of S-warfarin and Fe-O of CYP2C9, dose-dependent decrease in the glide scores (in silico). Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Hydrogen 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Hydrogen 67-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25823787-5 2015 CONCLUSION: CYP2C9*2 and CYP2C9*3 variants result in disruption of hydrogen bonding interactions with warfarin and longer distance between C7 and Fe-O thus impairing warfarin 7-hydroxylation due to lower binding affinity of warfarin. Warfarin 166-174 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25833162-4 2015 The results showed that MXC and HPTE inhibited both human CYP2C9 and rat liver CYP2C11 activity, with half-maximal inhibitory concentration (IC50) values of 15.47 +- 0.36 (MXC) and 8.87 +- 0.53 mumol/l (HPTE) for human CYP2C9, and of 22.45 +- 1.48 (MXC) and 24.63 +- 1.35 mumol/l (HPTE) for rat CYP2C11. 2,2-bis(4-hydroxyphenyl)-1,1,1-trichloroethane 32-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 25833162-4 2015 The results showed that MXC and HPTE inhibited both human CYP2C9 and rat liver CYP2C11 activity, with half-maximal inhibitory concentration (IC50) values of 15.47 +- 0.36 (MXC) and 8.87 +- 0.53 mumol/l (HPTE) for human CYP2C9, and of 22.45 +- 1.48 (MXC) and 24.63 +- 1.35 mumol/l (HPTE) for rat CYP2C11. 2,2-bis(4-hydroxyphenyl)-1,1,1-trichloroethane 32-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 219-225 25584932-4 2015 Black and Hispanic populations are known to have a higher prevalence of cardiovascular risk factors and disease, and a substantial proportion of black and Hispanic individuals possess genotypes of the cytochrome P450 (CYP) 2C9 enzyme involved in the metabolism of many NSAIDs and the CYP2D6 enzyme involved in metabolism of the dual opioid agonist/norepinephrine-serotonin reuptake inhibitor tramadol. Norepinephrine 348-362 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-226 26255664-0 2015 Identification and characterization of a novel CYP2C9 allelic variant in a warfarin-sensitive patient. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 26255664-6 2015 When expressed in yeast and insect cells, compared with wild-type enzyme, variant CYP2C9.60 exhibited lower protein expression capacity and showed significantly decreased metabolic activities for the hydroxylation of S-warfarin, tolbutamide and diclofenac. Warfarin 219-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 26255664-6 2015 When expressed in yeast and insect cells, compared with wild-type enzyme, variant CYP2C9.60 exhibited lower protein expression capacity and showed significantly decreased metabolic activities for the hydroxylation of S-warfarin, tolbutamide and diclofenac. Tolbutamide 229-240 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 26255664-6 2015 When expressed in yeast and insect cells, compared with wild-type enzyme, variant CYP2C9.60 exhibited lower protein expression capacity and showed significantly decreased metabolic activities for the hydroxylation of S-warfarin, tolbutamide and diclofenac. Diclofenac 245-255 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 26265036-7 2015 CONCLUSION: We confirmed the important role of VKORC1 and CYP2C9 polymorphisms in warfarin dose. Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 25584932-4 2015 Black and Hispanic populations are known to have a higher prevalence of cardiovascular risk factors and disease, and a substantial proportion of black and Hispanic individuals possess genotypes of the cytochrome P450 (CYP) 2C9 enzyme involved in the metabolism of many NSAIDs and the CYP2D6 enzyme involved in metabolism of the dual opioid agonist/norepinephrine-serotonin reuptake inhibitor tramadol. Serotonin 363-372 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-226 25584932-4 2015 Black and Hispanic populations are known to have a higher prevalence of cardiovascular risk factors and disease, and a substantial proportion of black and Hispanic individuals possess genotypes of the cytochrome P450 (CYP) 2C9 enzyme involved in the metabolism of many NSAIDs and the CYP2D6 enzyme involved in metabolism of the dual opioid agonist/norepinephrine-serotonin reuptake inhibitor tramadol. Tramadol 392-400 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-226 26827407-9 2015 DISCUSSION: The metabolism of warfarin involves several cytochrome P450 isoenzymes, including CYP1A2, CYP2C9, CYP2C19, and CYP3A4. Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 25282448-1 2015 The cytochrome P450 (CYP) 2C9 and vitamin K epoxide reductase complex 1 (VKORC1) genotypes have been strongly and consistently associated with warfarin dose requirements, and dosing algorithms incorporating genetic and clinical information have been shown to be predictive of stable warfarin dose. Warfarin 143-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-29 26053557-11 2015 A cocktail assay showed that CYP1A2, CYP2C9, CYP2C19, CYP2E1 and CYP3A4 can be inhibited by rutaecarpine in human liver microsomes. rutecarpine 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 25312789-5 2014 RESULTS: CYP2C9*2, CYP2C9*3, VKORC1-1639G>A, and APOE *epsilon4 were associated with lower warfarin doses. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 26422867-2 2015 We evaluated an existing warfarin dosing algorithm developed for Malaysian patients that was based only on VKORC1 and CYP2C9 genes. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 25372302-4 2014 In addition to the major metabolite 3"-OH-methyl-MFA, resulting from the benzylic hydroxylation by CYP2C9, 4"-hydroxy-MFA and 5-hydroxy-MFA were identified as metabolites resulting from oxidative metabolism of both aromatic rings of MFA. 3"-oh-methyl-mfa 36-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 25372302-4 2014 In addition to the major metabolite 3"-OH-methyl-MFA, resulting from the benzylic hydroxylation by CYP2C9, 4"-hydroxy-MFA and 5-hydroxy-MFA were identified as metabolites resulting from oxidative metabolism of both aromatic rings of MFA. Mefenamic Acid 49-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-105 25372302-6 2014 The major GSH conjugate was identified as 4"-OH-5"-glutathionyl-MFA and was formed at the highest activity by CYP1A2 and to a lesser extent by CYP2C9 and CYP3A4. Glutathione 10-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 25372302-6 2014 The major GSH conjugate was identified as 4"-OH-5"-glutathionyl-MFA and was formed at the highest activity by CYP1A2 and to a lesser extent by CYP2C9 and CYP3A4. 4"-oh-5"-glutathionyl-mfa 42-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 25224009-7 2014 Furthermore, VT-1161 weakly inhibited human CYP2C9, CYP2C19, and CYP3A4, suggesting a low drug-drug interaction potential. VT-1161 13-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 24891132-12 2014 The patients with CYP2C9*2 had a higher concentration of propofol at the time of LOC (p = 0.02). Propofol 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 25312789-5 2014 RESULTS: CYP2C9*2, CYP2C9*3, VKORC1-1639G>A, and APOE *epsilon4 were associated with lower warfarin doses. Warfarin 94-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 19-25 25312789-10 2014 CONCLUSION: CYP2C9*2, CYP2C9*3, VKORC1-1639G>A, and APOE *epsilon4 were associated with lower warfarin doses, while MDR1 3435C>T and UGT1A1(TA) n polymorphisms were associated with a requirement for higher doses. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25312789-10 2014 CONCLUSION: CYP2C9*2, CYP2C9*3, VKORC1-1639G>A, and APOE *epsilon4 were associated with lower warfarin doses, while MDR1 3435C>T and UGT1A1(TA) n polymorphisms were associated with a requirement for higher doses. Warfarin 97-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 25001883-2 2014 Previous studies have produced estimates of the effect of polymorphisms in Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) on stable warfarin dosing, but data on time in therapeutic range, initial dosing and adverse effects are limited. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-94 25001883-2 2014 Previous studies have produced estimates of the effect of polymorphisms in Cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase complex subunit 1 (VKORC1) on stable warfarin dosing, but data on time in therapeutic range, initial dosing and adverse effects are limited. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 25001883-7 2014 These data show novel associations between VKORC1-1639 and CYP2C9*2 and INR values in children taking warfarin, as well as replicating previous findings with regard to stable dose requirements. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 25951663-3 2014 The purpose of the present study was to clarify the role of 36 CYP2C9 alleles, 21 novel alleles (*36-*56) found in the Chinese population, in the oxidative metabolism of diclofenac in vitro. Diclofenac 170-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 25521356-4 2014 In the 214 (85.6%) cases with a major bleed 30 or more days after warfarin initiation, CYP2C9*3 was the only variant associated with bleeding (adjusted odds ratio: 2.05; 95% CI: 1.04, 4.04). Warfarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 25951663-8 2014 Thus, this study demonstrated that more attention should be given to subjects carrying these CYP2C9 alleles when administering diclofena. diclofena 127-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 25521356-5 2014 CONCLUSION: The CYP2C9*3 allele may double the risk of major bleeding among patients taking warfarin for 30 or more days. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25951663-0 2014 The role of CYP2C9 genetic polymorphisms in the oxidative metabolism of diclofenac in vitro. Diclofenac 72-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 25196644-5 2014 Both caffeic acid and quercetin were potent competitive inhibitors of CYP1A2 (Ki = 1.16 and 0.93 muM, respectively) and CYP2C9 (Ki = 0.95 and 1.67 muM, respectively). caffeic acid 5-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 25196644-5 2014 Both caffeic acid and quercetin were potent competitive inhibitors of CYP1A2 (Ki = 1.16 and 0.93 muM, respectively) and CYP2C9 (Ki = 0.95 and 1.67 muM, respectively). Quercetin 22-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 120-126 25361167-3 2014 The activities of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 were attenuated by EEP-B55 in a concentration-dependent manner, and artepillin C, kaempferide, dihydrokaempferide, isosakuranetin, and kaempferol were estimated to have potential for CYP inhibition. isosakuranetin 177-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 25427198-7 2014 The results suggested that MH was simultaneously a substrate and an inhibitor of CYP1A2 and CYP2C9, and MH had the potential to perpetrate drug-drug interactions with other CYP1A2 and CYP2C19 substrates. meranzin hydrate 27-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 25361167-3 2014 The activities of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 were attenuated by EEP-B55 in a concentration-dependent manner, and artepillin C, kaempferide, dihydrokaempferide, isosakuranetin, and kaempferol were estimated to have potential for CYP inhibition. eep-b55 81-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 25361167-3 2014 The activities of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 were attenuated by EEP-B55 in a concentration-dependent manner, and artepillin C, kaempferide, dihydrokaempferide, isosakuranetin, and kaempferol were estimated to have potential for CYP inhibition. kaempferol 197-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 25332267-2 2014 Bosentan is metabolized by the cytochrome CYP2C9 and inhibits the bile salt export pump, which is encoded by ABCB11. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 25361167-3 2014 The activities of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 were attenuated by EEP-B55 in a concentration-dependent manner, and artepillin C, kaempferide, dihydrokaempferide, isosakuranetin, and kaempferol were estimated to have potential for CYP inhibition. Dihydrokaempferide 157-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 25332267-3 2014 This suggests that genetic variants of CYP2C9 and/or ABCB11 may predispose patients to bosentan-induced liver injury. Bosentan 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 25332267-9 2014 CONCLUSION: Our data indicate hepatotoxicity of bosentan from decreased hepatic metabolism due to common variants of CYP2C9. Bosentan 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 25142093-0 2014 Influence of proton pump inhibitors and VKORC1 mutations on CYP2C9-mediated dose requirements of vitamin K antagonist therapy: a pilot study. Vitamin K 97-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 25142093-1 2014 Interindividual variations in dose requirements of oral vitamin K antagonists (VKA) are attributed to several factors, including genetic variant alleles of vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9), but also interaction with co-medications. Vitamin K 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 215-234 25142093-1 2014 Interindividual variations in dose requirements of oral vitamin K antagonists (VKA) are attributed to several factors, including genetic variant alleles of vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9), but also interaction with co-medications. Vitamin K 56-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 236-242 25099164-1 2014 Phenytoin is a widely used antiepileptic drug with a narrow therapeutic index and large interpatient variability, partly due to genetic variations in the gene encoding cytochrome P450 (CYP)2C9 (CYP2C9). Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-192 25099164-1 2014 Phenytoin is a widely used antiepileptic drug with a narrow therapeutic index and large interpatient variability, partly due to genetic variations in the gene encoding cytochrome P450 (CYP)2C9 (CYP2C9). Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 25099164-3 2014 We summarize evidence from the published literature supporting these associations and provide recommendations for the use of phenytoin based on CYP2C9 and/or HLA-B genotype (also available on PharmGKB: http://www.pharmgkb.org). Phenytoin 125-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 25142737-0 2014 Effects of cytochrome P450 2C9 polymorphism on bosentan metabolism. Bosentan 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-30 25142737-3 2014 The aim of this study was to assess the catalytic activities of 38 human CYP2C9 alleles, including 24 novel alleles (*36-*60) found in the Han Chinese population, toward bosentan (BOS) in vitro. Bosentan 170-178 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 25142737-3 2014 The aim of this study was to assess the catalytic activities of 38 human CYP2C9 alleles, including 24 novel alleles (*36-*60) found in the Han Chinese population, toward bosentan (BOS) in vitro. Bosentan 180-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 25142737-8 2014 These findings suggest that more attention should be directed to subjects carrying these infrequent CYP2C9 alleles when administering BOS in the clinic. Bosentan 134-137 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 100-106 25187484-0 2014 Mechanistic studies of the cationic binding pocket of CYP2C9 in vitro and in silico: metabolism of nonionizable analogs of tienilic acid. Ticrynafen 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24944083-7 2014 CYP2C9 and CYP2C19 inhibitors strongly inhibited R(+)-NAF metabolism, and CYP1A2, CYP2C8, CYP2D6 and CYP3A4/5 inhibitors moderately inhibited R(+)-NAF metabolism. r(+)-naf 49-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-130 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Ticrynafen 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Ticrynafen 15-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-130 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Ticrynafen 15-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Thiophenes 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-130 25187484-1 2014 Tienilic acid (TA) is selectively oxidized at the C-5 position of the thiophene ring by the human liver enzyme cytochrome P450 2C9 (CYP2C9). Thiophenes 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 25187484-3 2014 In this study, we investigated how chemical modification of TA influences the bioactivation by CYP2C9. Ticrynafen 60-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 24944083-7 2014 CYP2C9 and CYP2C19 inhibitors strongly inhibited R(+)-NAF metabolism, and CYP1A2, CYP2C8, CYP2D6 and CYP3A4/5 inhibitors moderately inhibited R(+)-NAF metabolism. r(+)-naf 142-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24944083-8 2014 CYP2C9 inhibitors strongly inhibited S(-)-NAF metabolism, and CYP2C8, CYP2C19 and CYP3A4/5 inhibitors moderately inhibited S(-)-NAF metabolism. naftopidil 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24944083-9 2014 Consistent with the results of chemical inhibitors experiments, recombinant human CYP2C9 and CYP2C19 contributed greatly to R(+)-NAF metabolism, and CYP2C9 contributed greatly to S(-)-NAF metabolism. r(+)-naf 124-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 24944083-9 2014 Consistent with the results of chemical inhibitors experiments, recombinant human CYP2C9 and CYP2C19 contributed greatly to R(+)-NAF metabolism, and CYP2C9 contributed greatly to S(-)-NAF metabolism. Sulfur 179-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 24944083-9 2014 Consistent with the results of chemical inhibitors experiments, recombinant human CYP2C9 and CYP2C19 contributed greatly to R(+)-NAF metabolism, and CYP2C9 contributed greatly to S(-)-NAF metabolism. naftopidil 180-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 149-155 24944083-11 2014 CYP2C9 plays the most important role in the demethylation and hydroxylation of both NAF enantiomers, CYP2C19 is another major CYP isoform that is involved in R(+)-NAF metabolism. naftopidil 84-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24944083-11 2014 CYP2C9 plays the most important role in the demethylation and hydroxylation of both NAF enantiomers, CYP2C19 is another major CYP isoform that is involved in R(+)-NAF metabolism. naftopidil 159-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 25280537-0 2014 Creatine kinase elevation caused by a combination of fluvastatin and telmisartan in a patient heterozygous for the CYP2C9*3 and ABCC2 -24C > T variants: a case report. Fluvastatin 53-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 25069408-2 2014 CYP2C9, VKROC1, and CYP4F2 play important roles in warfarin metabolism, and their genetic polymorphisms are related to the variability in dose determination. Warfarin 51-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24882266-2 2014 The main human CYP epoxygenases, i.e. CYP2C8, CYP2C9, CYP2C19 and CYP2J2, convert AA to four regioisomer epoxyeicosatrienoic acids (EETs). epoxyeicosatrienoic acids 105-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 24882266-2 2014 The main human CYP epoxygenases, i.e. CYP2C8, CYP2C9, CYP2C19 and CYP2J2, convert AA to four regioisomer epoxyeicosatrienoic acids (EETs). eets 132-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 24590863-6 2014 The standardized EJE was incubated with pooled human liver microsomes to assess the CYP2C9-, CYP2D6-, and CYP3A4-mediated metabolism of diclofenac, dextromethorphan, and testosterone, respectively. Diclofenac 136-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 24590863-9 2014 The selectivity of EJE for CYP2C9 rather than CYP3A4 and CYP2D6 led to perform the enzyme kinetics to explicate the mechanism underlying the inhibition of CYP2C9-mediated diclofenac 4"-hydroxylation. Diclofenac 171-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 24590863-9 2014 The selectivity of EJE for CYP2C9 rather than CYP3A4 and CYP2D6 led to perform the enzyme kinetics to explicate the mechanism underlying the inhibition of CYP2C9-mediated diclofenac 4"-hydroxylation. Diclofenac 171-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-161 25272989-4 2014 The reaction phenotype study showed that CYP3A4, CYP2C9, and CYP1A2 were the major cytochrome P450 isozymes in the metabolism of CSA. 3-hydroxy-4-prenyl-5-methoxystilbene-2-carboxylic acid 129-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 25272989-8 2014 CSA reversibly inhibited CYP3A4 and CYP2C9 activities in human liver microsomes with IC50 values of 28.3 and 31.3 muM, respectively, but exhibited no inhibition activities to CYP1A2, CYP2A6, CYP2C19, CYP2D6, and CYP2E1. 3-hydroxy-4-prenyl-5-methoxystilbene-2-carboxylic acid 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 25280537-0 2014 Creatine kinase elevation caused by a combination of fluvastatin and telmisartan in a patient heterozygous for the CYP2C9*3 and ABCC2 -24C > T variants: a case report. Telmisartan 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 24612223-4 2014 For example, SP 600125 was a potent inhibitor for CYP1A2, but enhanced the activity of CYP2C9 fourfolds. pyrazolanthrone 13-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-93 25079360-3 2014 Variation in warfarin pharmacokinetic and pharmacodynamic genes, such as CYP2C9 and VKORC1, do not fully explain the dose variability in African Americans. Warfarin 13-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 24750390-4 2014 It is known that variability in two genes (CYP2C9 and VKORC1) has a significant effect on individual response to warfarin dose. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 26052198-0 2014 Warfarin dose requirement in Turkish patients: the influences of patient characteristics and polymorphisms in CYP2C9, VKORC1 and factor VII. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 110-116 26052198-5 2014 RESULTS: The mean daily warfarin dosage was significantly higher in patients with the CYP2C9*1/*1 genotype than in the CYP2C9*2/*2 and CYP2C9*1/*3 groups (p <= 0.05). Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 26052198-5 2014 RESULTS: The mean daily warfarin dosage was significantly higher in patients with the CYP2C9*1/*1 genotype than in the CYP2C9*2/*2 and CYP2C9*1/*3 groups (p <= 0.05). Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 26052198-5 2014 RESULTS: The mean daily warfarin dosage was significantly higher in patients with the CYP2C9*1/*1 genotype than in the CYP2C9*2/*2 and CYP2C9*1/*3 groups (p <= 0.05). Warfarin 24-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 26052198-9 2014 CONCLUSIONS: Polymorphisms in CYP2C9, VKORC1 and factor VII did partially affect daily warfarin dose requirements, while age, gender, BMI and INR do not. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 25089947-1 2014 AIM: To develop an acenocoumarol (ACN) dosing algorithm for patients with atrial fibrillation or venous thromboembolism, considering the influence on the stable ACN dose of clinical factors and gene polymorphisms, including CYP2C9*2/*3, VKORC1, CYP4F2*3, ABCB1, APOE, CYP2C19*2/*17, and GGCX. Acenocoumarol 19-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-230 24663076-2 2014 In this study, we evaluate, for the first time, the influence of CYP2D6, CYP2C9 and ABCB1 genotypes on the steady-state plasma concentrations of fluoxetine and its active metabolite (S)-norfluoxetine, and on the clinical improvement in children and adolescent patients receiving fluoxetine treatment. Fluoxetine 145-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 25089947-4 2014 CONCLUSION: The variability in the stable ACN dose was better explained by a pharmacogenetic algorithm including clinical and genetic factors (CYP2C9, VKORC1, and APOE) than by a clinical algorithm, providing a more accurate dosage prediction. Acenocoumarol 42-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 25932358-10 2014 Our results indicated that the PCR-CTPP could be one of the alternative methods for genotyping VKORC1 G-1639A and CYP2C9 A1075C for Asians and Africans with similar allele frequencies to Japanese. ctpp 35-39 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 24779639-2 2014 In this article, the modulatory effects of extracts from vinegar-baked Radix Bupleuri (VBRB) and saikosaponins on the activity of CYP1A2, CYP2C9 and CYP3A4 were investigated in vitro. saikosaponin D 97-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 25932358-0 2014 A PCR method for VKORC1 G-1639A and CYP2C9 A1075C genotyping useful to warfarin therapy among Japanese. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 25182955-1 2014 BACKGROUND: Cytochrome P450 (CYP) 2C9 is expressed in the vascular endothelium and metabolizes arachidonic acid to biologically active epoxyeicosatrienoic acids (EETs), which have the crucial role in the modulation of cardiovascular homeostasis. Arachidonic Acid 95-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 25182955-1 2014 BACKGROUND: Cytochrome P450 (CYP) 2C9 is expressed in the vascular endothelium and metabolizes arachidonic acid to biologically active epoxyeicosatrienoic acids (EETs), which have the crucial role in the modulation of cardiovascular homeostasis. epoxyeicosatrienoic acids 135-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 25182955-1 2014 BACKGROUND: Cytochrome P450 (CYP) 2C9 is expressed in the vascular endothelium and metabolizes arachidonic acid to biologically active epoxyeicosatrienoic acids (EETs), which have the crucial role in the modulation of cardiovascular homeostasis. eets 162-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-37 24548191-6 2014 Fenofibrate was a potent in vitro inhibitor of CYP2C19 (IC50 = 0.2 mum) and CYP2B6 (IC50 = 0.7 mum) and a moderate inhibitor of CYP2C9 (IC50 = 9.7 mum). Fenofibrate 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 24919870-10 2014 In conclusion, VKORC1, CYP2C9*3, APOE and ABCB1 genotypes should be considered in prevention of overanticoagulation and bleeding events in the initiation of acenocoumarol therapy. Acenocoumarol 157-170 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 25088191-2 2014 A lead molecule was identified that has improved agonist efficacy relative to our clinical compound (APD597) and possesses reduced CYP2C9 inhibitory potential. APD597 101-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 24986093-0 2014 Drug-drug interaction of losartan and glimepiride metabolism by recombinant microsome CYP2C9*1, 2C9*3, 2C9*13, and 2C9*16 in vitro. Losartan 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 24986093-0 2014 Drug-drug interaction of losartan and glimepiride metabolism by recombinant microsome CYP2C9*1, 2C9*3, 2C9*13, and 2C9*16 in vitro. glimepiride 38-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 86-92 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-151 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 81-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-151 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). Losartan 81-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). glimepiride 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-151 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). glimepiride 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 161-167 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). glimepiride 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). glimepiride 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-2 2014 METHODS: In this study, we characterized the drug-drug interactions of losartan (LOS) and glimepiride (GLP) using recombinant cytochrome P450 (CYP) 2C9 enzymes (CYP2C9*1, CYP2C9*3, CYP2C9*13, and CYP2C9*16). glimepiride 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. Losartan 23-31 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. glimepiride 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. glimepiride 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. glimepiride 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-3 2014 RESULTS: Metabolism of losartan by recombinant CYP2C9* 1, CYP2C9*3, CYP2C9*13, and CYP2C9* 16 was inhibited by glimepiride competitively with IC50 values of 0.669 +- 0.055 microM, 0.424 +- 0.032 microM, 2.557 +- 0.058 microM, and 0.667 +- 0.039 microM, respectively. glimepiride 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24986093-4 2014 The inhibitory effect of glimepiride on losartan metabolism by CYP2C9*13 was marginal. glimepiride 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 24986093-5 2014 The apparent Ki value of glimepiride with CYP2C9*3 (0.0416 +- 0.0059 microM) was significantly lower than with CYP2C9*1 (0.1476 +- 0.0219 microM) and CYP2C9*16 (0.2671 +- 0.0456 microM). glimepiride 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 24986093-5 2014 The apparent Ki value of glimepiride with CYP2C9*3 (0.0416 +- 0.0059 microM) was significantly lower than with CYP2C9*1 (0.1476 +- 0.0219 microM) and CYP2C9*16 (0.2671 +- 0.0456 microM). glimepiride 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24986093-5 2014 The apparent Ki value of glimepiride with CYP2C9*3 (0.0416 +- 0.0059 microM) was significantly lower than with CYP2C9*1 (0.1476 +- 0.0219 microM) and CYP2C9*16 (0.2671 +- 0.0456 microM). glimepiride 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24986093-7 2014 The potencies for inhibition of glimepiride hydroxylation were determined to be CYP2C9*1~CYP2C9*3~CYP2C9*16 > CYP2C9*13 by 4 microM losartan. glimepiride 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 24986093-7 2014 The potencies for inhibition of glimepiride hydroxylation were determined to be CYP2C9*1~CYP2C9*3~CYP2C9*16 > CYP2C9*13 by 4 microM losartan. glimepiride 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 24986093-7 2014 The potencies for inhibition of glimepiride hydroxylation were determined to be CYP2C9*1~CYP2C9*3~CYP2C9*16 > CYP2C9*13 by 4 microM losartan. glimepiride 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 24986093-7 2014 The potencies for inhibition of glimepiride hydroxylation were determined to be CYP2C9*1~CYP2C9*3~CYP2C9*16 > CYP2C9*13 by 4 microM losartan. glimepiride 32-43 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. Losartan 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. glimepiride 131-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 25298588-0 2014 CYP2C9*3 polymorphism presenting as lethal subdural hematoma with low-dose warfarin. Warfarin 75-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. glimepiride 131-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 24986093-9 2014 CONCLUSIONS: Given these results, the potential inhibition of losartan metabolism by CYP2C9*3, CYP2C9*13, and CYP2C9*16 in vivo by glimepiride deserves further investigation. glimepiride 131-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 25246753-0 2014 Extremely elevated international normalized ratio of warfarin in a patient with CYP2C9*1/*3 and thyrotoxicosis. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 24730468-1 2014 OBJECTIVES: The aim of this study is to establish the inhibitory effects of 14 commonly used complementary and alternative medicines (CAM) on the metabolism of cytochrome P450 2C9 (CYP2C9) substrates 7-methoxy-4-trifluoromethyl coumarine (MFC) and tolbutamide. 7-methoxy-4-trifluoromethyl coumarine 200-237 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 24730468-1 2014 OBJECTIVES: The aim of this study is to establish the inhibitory effects of 14 commonly used complementary and alternative medicines (CAM) on the metabolism of cytochrome P450 2C9 (CYP2C9) substrates 7-methoxy-4-trifluoromethyl coumarine (MFC) and tolbutamide. mfc 239-242 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 24730468-1 2014 OBJECTIVES: The aim of this study is to establish the inhibitory effects of 14 commonly used complementary and alternative medicines (CAM) on the metabolism of cytochrome P450 2C9 (CYP2C9) substrates 7-methoxy-4-trifluoromethyl coumarine (MFC) and tolbutamide. Tolbutamide 248-259 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 181-187 24730468-5 2014 In human liver microsomes (HLM) the inhibition of CYP2C9-mediated metabolism of tolbutamide was determined, using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). Tolbutamide 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 25244877-9 2014 CONCLUSION: The association of CYP2C9 and VKORC1 genotypes and risks for adverse events due to exposure to warfarin was examined for the first time in Puerto Ricans. Warfarin 107-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 25317394-2 2014 Phenytoin, one of the common drugs implicated in gingival enlargement, is metabolized mainly by cytochrome P450 (CYP)2C9 and partly by CYP2C19. Phenytoin 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-120 25317394-4 2014 AIMS: The present study was undertaken to investigate the influence of the CYP2C9*2 and *3 variant genotypes on phenytoin hydroxylation in subjects diagnosed with epilepsy from South India, thus establishing the genetic polymorphisms leading to its defective hydroxylation process. Phenytoin 112-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 25410896-1 2014 The CYP2C9 c.449G>A (p.R150H, rs7900194) polymorphism, which confers the CYP2C9*8 allele, is common in persons of African descent and results in reduced clearance of the narrow therapeutic index drugs, warfarin and phenytoin. Warfarin 205-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 25410896-1 2014 The CYP2C9 c.449G>A (p.R150H, rs7900194) polymorphism, which confers the CYP2C9*8 allele, is common in persons of African descent and results in reduced clearance of the narrow therapeutic index drugs, warfarin and phenytoin. Warfarin 205-213 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 25410896-1 2014 The CYP2C9 c.449G>A (p.R150H, rs7900194) polymorphism, which confers the CYP2C9*8 allele, is common in persons of African descent and results in reduced clearance of the narrow therapeutic index drugs, warfarin and phenytoin. Phenytoin 218-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 25410896-1 2014 The CYP2C9 c.449G>A (p.R150H, rs7900194) polymorphism, which confers the CYP2C9*8 allele, is common in persons of African descent and results in reduced clearance of the narrow therapeutic index drugs, warfarin and phenytoin. Phenytoin 218-227 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 76-82 24908449-2 2014 Polymorphisms in two genes i.e. CYP2C9 (Cytochrome P450 2C9) and VKORC1 (Vitamin K epoxide reductase complex subunit 1) play a major role in warfarin dose variation and its related adverse effects. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 24908449-2 2014 Polymorphisms in two genes i.e. CYP2C9 (Cytochrome P450 2C9) and VKORC1 (Vitamin K epoxide reductase complex subunit 1) play a major role in warfarin dose variation and its related adverse effects. Warfarin 141-149 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-59 24910189-4 2014 As measured by liquid chromatography-electrospray ionization tandem mass spectrometry, ethambutol exhibited strong inhibitory potential against CYP1A2 and CYP2E1, moderate against CYP2C19 and CYP2D6 and weak against CYP2A6, CYP2C9 and CYP3A4, based on the IC50 values. Ethambutol 87-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 224-230 25042728-1 2014 INTRODUCTION: VKORC1 and cytochrome CYP2C9 genetic variants contribute largely to inter-individual variations in vitamin K antagonists (VKAs) dose requirements. Vitamin K 113-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 25042728-6 2014 Carriers of 2 and/or 3 variant alleles for CYP2C9 also required a lower dose of warfarin as compared with 1 1 carriers (35.0 [31.5-52.5] vs. 43.8 [35.0-60.2] mg/week, p=0.02; 35.0 [23.5-35.0] vs. 43.8 [35.0-60.2] mg/week, p<0.0001, respectively). Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 25202272-3 2014 The purpose of this study was to find out whether CYP2C9 was involved in this herb-herb interaction by using tolbutamide as a probe substrate in vivo and in vitro. Tolbutamide 109-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 25126975-3 2014 Only VKORC1 and CYP2C9 SNPs were observed to be significantly associated with warfarin dose. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 25096692-10 2014 Direct sequencing of CYP2C identified missense variant rs1057910 (CYP2C9*3) that showed significant association with phenytoin-related severe cutaneous adverse reactions (odds ratio, 12; 95% CI, 6.6-20; P=1.1 x 10(-17)). Phenytoin 117-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 24974237-0 2014 Prevalence of genetic polymorphisms of CYP2C9 and VKORC1 - implications for warfarin management and outcome in Croatian patients with acute stroke. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 24974237-1 2014 BACKGROUND: Data on the prevalence of CYP2C9 and VKORC1 genes and their influence on anticoagulant effect and warfarin dose in stroke patients are scarce. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 24974237-8 2014 Warfarin dosage complications were slightly more frequent among the carriers of CYP2C9 2, 3 compared to the carriers of VKORC1 1173T alleles (68. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 25096692-11 2014 The statistically significant association between CYP2C9*3 and phenytoin-related severe cutaneous adverse reactions was observed in additional samples from Taiwan, Japan, and Malaysia. Phenytoin 63-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 25096692-12 2014 A meta-analysis using the data from the 3 populations showed an overall odds ratio of 11 (95% CI, 6.2-18; z=8.58; P < .00001) for CYP2C9*3 association with phenytoin-related severe cutaneous adverse reactions. Phenytoin 159-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 133-139 25096692-13 2014 Delayed clearance of plasma phenytoin was detected in patients with severe cutaneous adverse reactions, especially CYP2C9*3 carriers, providing a functional link of the associated variants to the disease. Phenytoin 28-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 25096692-14 2014 CONCLUSIONS AND RELEVANCE: This study identified CYP2C variants, including CYP2C9*3, known to reduce drug clearance, as important genetic factors associated with phenytoin-related severe cutaneous adverse reactions. Phenytoin 162-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 25014006-2 2014 Herein, a nanotube array enzymatic reactor (CYP2C9/Au/TNA) was constructed by electrostatically adsorbing enzyme on the inner wall of TiO2 nanotube arrays (TNAs). titanium dioxide 134-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 25014006-5 2014 The cytochrome P450 2C9 enzyme (CYP2C9) was confined inside TNAs as a model. tnas 60-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-23 25014006-5 2014 The cytochrome P450 2C9 enzyme (CYP2C9) was confined inside TNAs as a model. tnas 60-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 24797541-0 2014 Frequency of CYP2C9 and VKORC1 gene polymorphisms and their influence on warfarin dose in Egyptian pediatric patients. Warfarin 73-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24830941-2 2014 Initial studies using primary human hepatocytes showed induction of both the CYP2C9 and CYP2C19 genes by tert-butylhydroquinone (tBHQ). 2-tert-butylhydroquinone 105-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 24830941-2 2014 Initial studies using primary human hepatocytes showed induction of both the CYP2C9 and CYP2C19 genes by tert-butylhydroquinone (tBHQ). 2-tert-butylhydroquinone 129-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 77-83 24830941-6 2014 Using specific kinase inhibitors for mitogen-activated protein kinase, we showed that extracellular signal-regulated kinase and Jun N-terminal kinase are essential for tBHQ-induced expression of CYP2C9. 2-tert-butylhydroquinone 168-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 24322786-0 2014 High warfarin sensitivity in carriers of CYP2C9*35 is determined by the impaired interaction with P450 oxidoreductase. Warfarin 5-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 24322786-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes many clinically important drugs including warfarin and diclofenac. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24322786-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes many clinically important drugs including warfarin and diclofenac. Warfarin 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24322786-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes many clinically important drugs including warfarin and diclofenac. Diclofenac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24322786-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes many clinically important drugs including warfarin and diclofenac. Diclofenac 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24322786-2 2014 We have recently reported a new allelic variant, CYP2C9*35, found in a warfarin hypersensitive patient with Arg125Leu and Arg144Cys mutations. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Warfarin 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Warfarin 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Warfarin 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Warfarin 105-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Diclofenac 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Diclofenac 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Diclofenac 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. Diclofenac 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. NADP 134-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. NADP 134-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. NADP 134-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-4 2014 CYP2C9.1 and CYP2C9-Arg144Cys expressed in human embryonic kidney 293 cells effectively metabolized both S-warfarin and diclofenac in NADPH-dependent reactions, whereas CYP2C9-Arg125Leu or CYP2C9.35 were catalytically silent. NADP 134-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24322786-5 2014 However, when NADPH was replaced by a direct electron donor to CYPs, cumene hydroperoxide, hereby bypassing the CYP oxidoreductase (POR), all variant enzymes were active, indicating unproductive interactions between CYP2C9.35 and POR. NADP 14-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 216-222 24322786-5 2014 However, when NADPH was replaced by a direct electron donor to CYPs, cumene hydroperoxide, hereby bypassing the CYP oxidoreductase (POR), all variant enzymes were active, indicating unproductive interactions between CYP2C9.35 and POR. cumene hydroperoxide 69-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 216-222 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. NADP 143-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. NADP 143-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. NADP 143-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. Warfarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. Warfarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 24322786-7 2014 In conclusion, our data strongly suggest that the Arg125Leu mutation in CYP2C9.35 prevents CYP2C9-POR interactions resulting in the absence of NADPH-dependent CYP2C9-catalyzed activity in vivo, thus influencing the warfarin sensitivity in the carriers of this allele. Warfarin 215-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 25303293-0 2014 Relationship between the CYP2C9 IVS8-109A>T polymorphism and high losartan hydroxylation in healthy Ecuadorian volunteers. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 25003537-0 2014 Impact of CYP2C9*3/*3 genotype on the pharmacokinetics and pharmacodynamics of oxicam NSAIDs. oxicam 79-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 25003538-0 2014 Response to Suarez-Kurtz"s comments on strongly increased exposure of meloxicam in CYP2C9*3/*3 individuals. Meloxicam 70-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 25303293-1 2014 AIM: The CYP2C9 IVS8-109T allele was recently found to be more frequent among Swedish individuals, who have the highest losartan metabolic ratio (MR; losartan:E-3174). Losartan 120-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 25303293-1 2014 AIM: The CYP2C9 IVS8-109T allele was recently found to be more frequent among Swedish individuals, who have the highest losartan metabolic ratio (MR; losartan:E-3174). Losartan 150-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 9-15 25303293-2 2014 Thus, the influence of the CYP2C9 IVS8-109A>T polymorphism on the losartan MR was evaluated among healthy Ecuadorians. Losartan 69-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 25303293-7 2014 Further investigation needs to be carried out in order to clarify the relevance of the SNP of CYP2C9 IVS8-109A>T on losartan hydroxylation across populations and its potential implications in CYP2C9 activity. Losartan 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 25303293-7 2014 Further investigation needs to be carried out in order to clarify the relevance of the SNP of CYP2C9 IVS8-109A>T on losartan hydroxylation across populations and its potential implications in CYP2C9 activity. Losartan 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 195-201 25197954-0 2014 Inhibition of cytochrome P450 2C9 expression and activity in vitro by allyl isothiocyanate. allyl isothiocyanate 70-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-33 25197954-4 2014 In this study, we found that allyl isothiocyanate reduced catalytic activity, messenger ribonucleic acid, and protein expression of cytochrome P450 2C9 in HepaRG cells. allyl isothiocyanate 29-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-151 25197954-5 2014 An investigation of the transcriptional activity of the pregnane X receptor and the constitutive androstane receptor revealed that allyl isothiocyanate disrupted the transcriptional coregulation effects of the pregnane X receptor/constitutive androstane receptor with several important coregulators and interfered with the assembly of transcriptional complexes of the cytochrome P450 2C9 pregnane X receptor/constitutive androstane receptor-response element. isothiocyanic acid 137-151 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 368-387 25197954-6 2014 The decrease of cytochrome P450 2C9 expression and activity mediated by allyl isothiocyanate suggested that this agent could alter the metabolism of drugs metabolized by cytochrome P450 2C9. allyl isothiocyanate 72-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-35 25197954-6 2014 The decrease of cytochrome P450 2C9 expression and activity mediated by allyl isothiocyanate suggested that this agent could alter the metabolism of drugs metabolized by cytochrome P450 2C9. allyl isothiocyanate 72-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 170-189 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 63-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-109 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 63-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 63-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25117329-6 2014 We were able to correctly infer genotypes for the warfarin-related loci VKORC1 and CYP2C9 alleles 2, 3, 5, and 11 and also clopidogrel-related CYP2C19 alleles 2 and 17 for a small sample of Brazilian individuals, as well as for HapMap samples. Warfarin 50-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 219-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-109 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 219-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24821498-1 2014 On the basis of the photo-induced electron transfer (PET) from CdTe quantum dots (QDs) to cytochrome P450 2C9 (CYP2C9), a light-controlled drug metabolism system was successfully designed by using CYP2C9 functionalized-CdTe QDs as photocatalysts. cadmium telluride 219-223 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 24794636-0 2014 Structural and energetic analysis to provide insight residues of CYP2C9, 2C11 and 2E1 involved in valproic acid dehydrogenation selectivity. Valproic Acid 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 24794636-2 2014 In this study, both methods were combined to predict the regioselectivity in the binding mode of valproic acid (VPA) on three cytochrome P-450 (CYP) isoforms CYP2C9, CYP2C11, and CYP2E1, which are involved in the biotransformation of VPA yielding reactive hepatotoxic intermediate 2-n-propyl-4-pentenoic acid (4nVPA). Valproic Acid 97-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 24794636-2 2014 In this study, both methods were combined to predict the regioselectivity in the binding mode of valproic acid (VPA) on three cytochrome P-450 (CYP) isoforms CYP2C9, CYP2C11, and CYP2E1, which are involved in the biotransformation of VPA yielding reactive hepatotoxic intermediate 2-n-propyl-4-pentenoic acid (4nVPA). Valproic Acid 112-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 25221392-4 2014 We hypothesize that the polymorphisms in ABCB1, Cyp2C9, Cyp2C19 and methylene tetrahydrofolate reductase (MTHFR) might result in differential expression resulting in differential drug transport, drug metabolism and folate metabolism, which in turn may contribute to the teratogenic impact of AEDs. Folic Acid 88-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 24839071-4 2014 In vitro, glyburide is metabolized (intrinsic clearance, 52.9 muL/min/mg-microsomal protein) by CYP3A4, CYP2C9, and CYP2C8 with fraction metabolism of 0.53, 0.36, and 0.11, respectively. Glyburide 10-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 104-110 24760401-3 2014 The objective of this study was to evaluate the effect of IPE on the pharmacokinetic and anticoagulation pharmacodynamics of warfarin, a substrate of cytochrome P450 2C9-mediated metabolism. eicosapentaenoic acid ethyl ester 58-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-169 24590592-9 2014 Since other sulfonylureas in clinical use are substrates of CYP2C9, their hepatic CLint changes have the potential to cause clinically relevant pharmacokinetic changes in a hyperlipidemic state. Sulfonylurea Compounds 12-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 24942396-1 2014 Observational studies have overwhelmingly shown that variants in the genes CYP2C9 and VKORC1 are significant determinants of individual dose of coumarin anticoagulants needed to maintain a therapeutic international normalized ratio (INR). coumarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 24760401-3 2014 The objective of this study was to evaluate the effect of IPE on the pharmacokinetic and anticoagulation pharmacodynamics of warfarin, a substrate of cytochrome P450 2C9-mediated metabolism. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-169 24768782-1 2014 BACKGROUND: Cyclophosphamide (CP), an alkylating chemotherapeutic drug, is catalyzed by the Phase I cytochrome P450 (CYPs) isozymes - CYP3A4, CYP3A5, CYP2B6, CYP2C8, CYP2C9 and CYP2C19. Cyclophosphamide 12-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 25155935-0 2014 Genetic epidemiology of pharmacogenetic variations in CYP2C9, CYP4F2 and VKORC1 genes associated with warfarin dosage in the Indian population. Warfarin 102-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24757038-2 2014 Voriconazole, a second-generation triazole antifungal agent, is both a substrate and potent inhibitor of CYP isoenzymes, specifically CYP2C19, CYP2C9, and CYP3A4; thus, the potential for drug-drug interactions with voriconazole is high. Voriconazole 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 24594507-5 2014 The hepatic cytochrome P450 (CYP) 2C protein expression level in the menthol administration group was significantly increased compared to that in the control group. Menthol 69-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-36 25136521-0 2014 Association of cytochrome P450 2C9 polymorphism with locally advanced head and neck squamous cell carcinoma and response to concurrent cisplatin-based radical chemoradiation. Cisplatin 135-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-34 25136521-1 2014 AIMS: The aim of the present study is to investigate the association between polymorphism of cytochrome P450 2C9 (CYP2C9) enzyme with head and neck squamous cell carcinoma (HNSCC) and response in patients receiving cisplatin-based radical chemoradiation (CT-RT). Cisplatin 215-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-112 25136521-1 2014 AIMS: The aim of the present study is to investigate the association between polymorphism of cytochrome P450 2C9 (CYP2C9) enzyme with head and neck squamous cell carcinoma (HNSCC) and response in patients receiving cisplatin-based radical chemoradiation (CT-RT). Cisplatin 215-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 25136521-5 2014 Tobacco intake in the form of chewing or smoking and alcohol intake resulted in several folds increase in the risk to HNSCC in the cases carrying variant genotypes of CYP2C9*2 or CYP2C9*013. Alcohols 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 167-173 25136521-5 2014 Tobacco intake in the form of chewing or smoking and alcohol intake resulted in several folds increase in the risk to HNSCC in the cases carrying variant genotypes of CYP2C9*2 or CYP2C9*013. Alcohols 53-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 179-185 25136521-6 2014 Further, majority of the cases assessed for response (n = 436) carrying variant alleles of CYP2C9*2 (69.6%) or CYP2C9*3 (65.2%) were found to respond poorly to cisplatin-based radical CT-RT. Cisplatin 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 25136521-6 2014 Further, majority of the cases assessed for response (n = 436) carrying variant alleles of CYP2C9*2 (69.6%) or CYP2C9*3 (65.2%) were found to respond poorly to cisplatin-based radical CT-RT. Cisplatin 160-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24601977-2 2014 We performed a retrospective, cross-sectional study to examine the effect of variant CYP2C9 and VKORC1 genotypes on warfarin dose in 100 children. Warfarin 116-124 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24594507-7 2014 The addition of menthol to human hepatic cells, HepaRG cells, caused an increase in the mRNA expression level of CYP2C9. Menthol 16-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 23800980-2 2014 CYP2C9 and VKORC1 are two major genetic factors associated with inter-individual and inter-ethnic variability in the warfarin dose. Warfarin 117-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24842639-0 2014 CYP2C9, SLCO1B1, SLCO1B3, and ABCB11 polymorphisms in patients with bosentan-induced liver toxicity. Bosentan 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24842639-4 2014 We conducted a nested case-control study to further explore the relationship between functional polymorphisms of gene products involved in bosentan pharmacokinetics (OATP1B1, OATP1B3, and CYP2C9) or hepatobiliary transporters affected by bosentan (ABCB11) and bosentan-induced liver toxicity. Bosentan 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 188-194 23800980-3 2014 AIM: This study aims to assess the impact of VKORC1-1639G>A polymorphism and the most common CYP2C9 variant alleles (*2 and *3) on warfarin response in Egyptian patients. Warfarin 134-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 23800980-7 2014 RESULTS: Patients carrying VKORC1 and CYP2C9 variant genotypes needed a 44.8 % lower mean daily warfarin dose as compared to wild types. Warfarin 96-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 23800980-10 2014 Multiple regression analysis showed that, VKORC1-1639G>A, age, CYP2C9*3, and smoking status explained 43.4 % of the overall variability in the warfarin dose. Warfarin 146-154 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 23800980-11 2014 CONCLUSION: VKORC1-1639G>A and CYP2C9 polymorphisms contribute to the difference in warfarin dose requirements and quality of anticoagulation amongst Egyptian patients. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 24602049-2 2014 This study aimed to evaluate the effect of gene polymorphisms of CYP2C9, VKORC1, thrombomodulin (THBD) and C-reactive protein (CRP) on the risk of bleeding complications of warfarin at therapeutic INR in Korean patients with mechanical cardiac valves. Warfarin 173-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 24612117-10 2014 WHAT IS NEW AND CONCLUSION: The patient required a high warfarin dose because of the VKORC1 genotype, and induction of CYP2C9 by carbamazepine. Carbamazepine 129-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 119-125 24574079-9 2014 ANN analysis identified several predictors of warfarin dosage including race; age; height; weight; cytochrome P450 (CYP)2C9 genotype; VKORC1 genotype; sulfonamide, azole antifungals, or macrolide administration; carbamazepine, phenytoin, or rifampicin administration; and amiodarone administration. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 99-123 24474498-0 2014 VKORC1 and CYP2C9 genotypes are predictors of warfarin-related outcomes in children. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24474498-9 2014 CYP2C9*3 carriers were also at increased risk of major bleeding while receiving warfarin (adjusted OR = 11.28). Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24474498-10 2014 An additional variant in CYP2C9 (rs7089580) was significantly associated with warfarin dose (P = 0.020) in a multivariate clinical and genetic model. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 25-31 24474498-11 2014 CONCLUSIONS: This study confirms the importance of VKORC1/CYP2C9 genotypes for warfarin dosing in a young pediatric cohort and demonstrates an impact of genetic factors on clinical outcomes in children. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 24474498-12 2014 Furthermore, we identified an additional variant in CYP2C9 of potential relevance for warfarin dosing in children. Warfarin 86-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 24018621-1 2014 Using a derivation cohort (N=349), we developed the first warfarin dosing algorithm that includes recently discovered polymorphisms in VKORC1 and CYP2C9 associated with warfarin dose requirement in African Americans (AAs). Warfarin 58-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 24018621-1 2014 Using a derivation cohort (N=349), we developed the first warfarin dosing algorithm that includes recently discovered polymorphisms in VKORC1 and CYP2C9 associated with warfarin dose requirement in African Americans (AAs). Warfarin 169-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 146-152 24165757-12 2014 These results suggest that CYP2C9 has a role in the oxidation reactions of THT and indicate that it may be possible to predict the efficacy of Bu-based myeloablative conditioning before HSCT on the basis of CYP genotypes and Bu MRs. tetrahydrothiophene 75-78 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 24165757-12 2014 These results suggest that CYP2C9 has a role in the oxidation reactions of THT and indicate that it may be possible to predict the efficacy of Bu-based myeloablative conditioning before HSCT on the basis of CYP genotypes and Bu MRs. Busulfan 143-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Simvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Simvastatin 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Atorvastatin 13-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Atorvastatin 13-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Lovastatin 30-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Lovastatin 30-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Fluvastatin 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 24974574-4 2014 Simvastatin, atorvastatin and lovastatin are metabolized by CYP3A4, fluvastatin by CYP2C9, while rosuvastatin by CYP2C9 and 2C19. Rosuvastatin Calcium 97-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-119 24928022-6 2014 RESULTS: VKORC1 and CYP2C9 gene polymorphisms could explain more than 50% Warfarin maintenance dose variation in recruited patients, while CYP4F2 gene polymorphisms could only explain 1%. Warfarin 74-82 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 20-26 24928022-8 2014 VKORC1 and CYP2C9 gene polymorphisms have a greater impact on Warfarin maintenance dose compared with demographic characteristics and concomitant medications. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24928022-9 2014 CONCLUSION: VKORC1 and CYP2C9 gene polymorphisms have a significant impact on Warfarin maintenance dose in Chinese Han population. Warfarin 78-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 24956244-0 2014 Warfarin dose requirements in a patient with the CYP2C9*14 allele. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 24799042-4 2014 The device enables users to manipulate the expression of individual and multiple human metabolizing-enzyme genes (such as CYP3A4, CYP2D6, CYP2C9, CYP1A2, CYP2E1 and UGT1A4) in THLE-2 cell microarrays. thle 176-180 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 24802986-3 2014 In this study, the in vitro inhibitory effects of scutellarin on six major human CYPs (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) and six rat CYPs (CYP1A2, CYP2C7, CYP2C11, CYP2C79, CYP2D4, and CYP3A2) activities were examined by using liquid chromatography-tandem mass spectrometry. scutellarin 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 24550229-0 2014 1-Aminobenzotriazole coincubated with (S)-warfarin results in potent inactivation of CYP2C9. 1-aminobenzotriazole 0-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24550229-0 2014 1-Aminobenzotriazole coincubated with (S)-warfarin results in potent inactivation of CYP2C9. Warfarin 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24550229-3 2014 Here we propose that ABT preferentially binds to the warfarin-binding pocket in the CYP2C9 active-site cavity; thus, ABT bioactivation and subsequent inactivation is not favored. 1-aminobenzotriazole 21-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 24550229-3 2014 Here we propose that ABT preferentially binds to the warfarin-binding pocket in the CYP2C9 active-site cavity; thus, ABT bioactivation and subsequent inactivation is not favored. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 24550229-4 2014 Therefore, coincubation with (S)-warfarin would result in displacement of ABT from the warfarin-binding pocket and subsequent binding to the active site, converting ABT into a potent inactivator of CYP2C9. Warfarin 29-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 24550229-4 2014 Therefore, coincubation with (S)-warfarin would result in displacement of ABT from the warfarin-binding pocket and subsequent binding to the active site, converting ABT into a potent inactivator of CYP2C9. 1-aminobenzotriazole 74-77 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 24550229-4 2014 Therefore, coincubation with (S)-warfarin would result in displacement of ABT from the warfarin-binding pocket and subsequent binding to the active site, converting ABT into a potent inactivator of CYP2C9. Warfarin 33-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 24550229-4 2014 Therefore, coincubation with (S)-warfarin would result in displacement of ABT from the warfarin-binding pocket and subsequent binding to the active site, converting ABT into a potent inactivator of CYP2C9. 1-aminobenzotriazole 165-168 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 198-204 24550229-5 2014 To test this hypothesis, in vitro studies were conducted using various coincubation combinations of ABT and (S)-warfarin or diclofenac to modulate the effectiveness of CYP2C9 inactivation by ABT. 1-aminobenzotriazole 100-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 24550229-5 2014 To test this hypothesis, in vitro studies were conducted using various coincubation combinations of ABT and (S)-warfarin or diclofenac to modulate the effectiveness of CYP2C9 inactivation by ABT. Warfarin 108-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 24550229-5 2014 To test this hypothesis, in vitro studies were conducted using various coincubation combinations of ABT and (S)-warfarin or diclofenac to modulate the effectiveness of CYP2C9 inactivation by ABT. Diclofenac 124-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 24550229-5 2014 To test this hypothesis, in vitro studies were conducted using various coincubation combinations of ABT and (S)-warfarin or diclofenac to modulate the effectiveness of CYP2C9 inactivation by ABT. 1-aminobenzotriazole 191-194 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 24550229-7 2014 The kinetic parameters of time-dependent inhibition of ABT for CYP2C9 in the absence and presence of (S)-warfarin (20 muM) were 0.0826 and 0.273 min(-1) for kinact and 3.49 and 0.157 mM for KI, respectively. 1-aminobenzotriazole 55-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 24550229-7 2014 The kinetic parameters of time-dependent inhibition of ABT for CYP2C9 in the absence and presence of (S)-warfarin (20 muM) were 0.0826 and 0.273 min(-1) for kinact and 3.49 and 0.157 mM for KI, respectively. Warfarin 101-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 24956244-6 2014 This report adds to the limited data of the effects of the CYP2C9*14 allele on warfarin dose requirements. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 24530212-0 2014 Influence of CYP2C9 polymorphism and phenytoin co-administration on acenocoumarol dose in patients with cerebral venous thrombosis. Acenocoumarol 68-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 24530212-6 2014 During the initiation phase of anticoagulation the CYP2C9*2 allele was independently associated with low acenocoumarol dose requirement (Adjusted OR 5.38; 95%CI 1.65-17.49; p=0.005). Acenocoumarol 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 24530212-12 2014 CONCLUSION: This study emphasizes the fact that polymorphisms in CYP2C9 gene and co-medication with phenytoin alter the anticoagulant effect of acenocoumarol. Phenytoin 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 24530212-12 2014 CONCLUSION: This study emphasizes the fact that polymorphisms in CYP2C9 gene and co-medication with phenytoin alter the anticoagulant effect of acenocoumarol. Acenocoumarol 144-157 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 24165884-7 2014 These data suggest that etravirine is a weak CYP3A isozyme inducer and minimally inhibits CYP2C9, 2C19, and P-glycoprotein activity. etravirine 24-34 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 90-96 25042915-9 2014 CONCLUSION: Our algorithm based on VKORC1, CYP2C9 and CYP4F2 polymorphisms can help to predict the warfarin maintenance dose in Chinese Han Population. Warfarin 99-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 24966969-1 2014 Single nucleotide polymorphisms (SNPs) of VKORC (1173T/C, rs9934438) and CYP2C9 (1075A/C, rs1057910) are major contributory factors on the sensitivity of warfarin in Chinese. Warfarin 154-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 73-79 24118918-0 2014 In vitro assessment of 36 CYP2C9 allelic isoforms found in the Chinese population on the metabolism of glimepiride. glimepiride 103-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 24118918-9 2014 This study provided the most comprehensive data on the enzymatic activities of all reported CYP2C9 variants in the Chinese population with regard to the commonly used antidiabetic drug, glimepiride. glimepiride 186-197 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 24118918-10 2014 Our results indicate that most of the tested rare alleles significantly decrease the catalytic activity of CYP2C9 variants towards glimepiride hydroxylation in vitro. glimepiride 131-142 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 107-113 24224579-3 2014 Phenprocoumon metabolism is mediated by CYP2C9 and CYP3A enzymes. Phenprocoumon 0-13 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 24519941-5 2014 Sulfaphenazole (SFZ), a selective inhibitor of human cytochrome P450 (CYP) 2C9 isozyme, was identified as a novel and leading cytoprotective compound. Sulfaphenazole 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-78 24337438-9 2014 In conclusion, in addition to body-weight and amiodarone-intake, pharmacogenetic factors (VKORC1, CYP4F2, ABCB1) related to the pharmacodynamic effect and transport of fluindione significantly influenced the dose requirement in elderly patients while CYP2C9 did not. fluindione 168-178 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 251-257 24337438-10 2014 Studies are required to know whether fluindione could be an alternative VKA in carriers of polymorphic CYP2C9 alleles, hypersensitive to coumarins. fluindione 37-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 24337438-10 2014 Studies are required to know whether fluindione could be an alternative VKA in carriers of polymorphic CYP2C9 alleles, hypersensitive to coumarins. Coumarins 137-146 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 103-109 24723938-1 2014 CYP2C8 and CYP2C9 are involved in the inactivation of several non-steroidal anti-inflammatory drugs, including ibuprofen. Ibuprofen 111-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24519941-5 2014 Sulfaphenazole (SFZ), a selective inhibitor of human cytochrome P450 (CYP) 2C9 isozyme, was identified as a novel and leading cytoprotective compound. Sulfaphenazole 16-19 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-78 24342125-4 2014 Isoxanthohumol from hops was the most potent inhibitor of CYP2C8 with an IC50 of 0.2 muM, whereas 8-prenylnaringenin was the most potent inhibitor of CYP1A2, CYP2C9 and CYP2C19 with IC50 values of 1.1 muM, 1.1 muM and 0.4 muM, respectively. 8-prenylnaringenin 98-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 24593903-1 2014 BACKGROUND: Two variants in the gene encoding the cytochrome P450 2C9 enzyme (CYP2C9) are considered the most significant genetic risk factors associated with bleeding after warfarin prescription. Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-69 24593903-0 2014 Genetic polymorphisms in warfarin and tacrolimus-related genes VKORC1, CYP2C9 and CYP3A5 in the Greek-Cypriot population. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 24593903-0 2014 Genetic polymorphisms in warfarin and tacrolimus-related genes VKORC1, CYP2C9 and CYP3A5 in the Greek-Cypriot population. Tacrolimus 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 24594849-2 2014 Isozyme CYP2C9 is reported to be inhibited by benzbromarone (BzBr) and this phenomenon was hitherto explained by classical active-site binding. Benzbromarone 46-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 24594849-2 2014 Isozyme CYP2C9 is reported to be inhibited by benzbromarone (BzBr) and this phenomenon was hitherto explained by classical active-site binding. Benzbromarone 61-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 24594849-5 2014 In silico docking of various BzBr analogs with known crystal structure of CYP2C9 did not provide any evidence in support of active-site based inhibition hypothesis. Benzbromarone 29-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 24594849-10 2014 Further, amphipathic derivatives of vitamins C & E (scavengers of diffusible reactive oxygen species or DROS) effectively inhibited CYP2C9 reactions in different reaction setups. vitamins c & 36-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 24594849-10 2014 Further, amphipathic derivatives of vitamins C & E (scavengers of diffusible reactive oxygen species or DROS) effectively inhibited CYP2C9 reactions in different reaction setups. Reactive Oxygen Species 81-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 24594849-10 2014 Further, amphipathic derivatives of vitamins C & E (scavengers of diffusible reactive oxygen species or DROS) effectively inhibited CYP2C9 reactions in different reaction setups. dros 108-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 24357089-6 2014 The PG/PK study of indapamide demonstrated that the polymorphic SNPs CYP2C9 rs4918758 and CYP2C19 rs4244285 appeared to confer lowered enzyme activity, as indicated by increased C max (25% ~ 64%), increased area under the plasma level-time curves (30~76%), increased area under the time infinity (43% ~ 80%), and lower apparent clearance values than PK for wild-type indapamide. Indapamide 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 24357089-6 2014 The PG/PK study of indapamide demonstrated that the polymorphic SNPs CYP2C9 rs4918758 and CYP2C19 rs4244285 appeared to confer lowered enzyme activity, as indicated by increased C max (25% ~ 64%), increased area under the plasma level-time curves (30~76%), increased area under the time infinity (43% ~ 80%), and lower apparent clearance values than PK for wild-type indapamide. Indapamide 367-377 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 24357089-7 2014 Our results reinforce the biochemical support of CYP2C19 in indapamide metabolism and identify a possible new participating enzyme CYP2C9. Indapamide 60-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 131-137 24593903-1 2014 BACKGROUND: Two variants in the gene encoding the cytochrome P450 2C9 enzyme (CYP2C9) are considered the most significant genetic risk factors associated with bleeding after warfarin prescription. Warfarin 174-182 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 24387348-5 2014 Current evidence suggests that garlic and ginkgo can be safely co-administered, whereas CYP2C9 substrates (e.g., warfarin) should be monitored closely when ginseng therapy is started. Warfarin 113-121 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 24464553-0 2014 Characterizing the effect of cytochrome P450 (CYP) 2C8, CYP2C9, and CYP2D6 genetic polymorphisms on stereoselective N-demethylation of fluoxetine. Fluoxetine 135-145 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 24464553-6 2014 Five CYP2C9 variants and CYP2D6.1 exhibited significantly stereoselective kinetic profiles prior to R-FLX, and CYP2C8.3 showed a slight stereoselectivity. r-flx 100-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 5-11 24464553-8 2014 Together, these findings suggest that CYP2C9 and CYP2D6 polymorphism may play an important role in the clearance of FLX and also in the stereoselective kinetic profiles of FLX enantiomers. Fluoxetine 116-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 24322971-4 2014 Nevertheless, VKORC1, CYP2C9, and CYP4F2 variants have been associated with warfarin dose requirements, and CYP2C19 variants have been associated with perturbed antiplatelet response to clopidogrel. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 22-28 24660123-1 2014 Warfarin is the most common anticoagulant prescribed and its metabolism has been linked to two specific genes, CYP2C9 and VKORC1. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 24368832-2 2014 Levonorgestrel, a commonly employed progestin component of combined oral contraceptives, was implicated in drug-drug interactions mediated via CYP2C9. Levonorgestrel 0-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 24368832-4 2014 In the current study, we examined the phenotypic status of CYP2C9 using low-dose (125 mg) tolbutamide before and after oral contraceptive use in reproductive age women. Tolbutamide 90-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 24368832-6 2014 The natural logarithm of tolbutamide C24, a metric for CYP2C9 phenotype, was found to be equivalent (within 80%-125% equivalency boundaries) before and after oral contraceptive use. Tolbutamide 25-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 23932726-0 2014 Frequency of CYP2C9 polymorphisms in Polynesian people and potential relevance to management of gout with benzbromarone. Benzbromarone 106-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 23932726-2 2014 The uricosuric drug benzbromarone is an alternative treatment but CYP2C9 poor metabolisers (PMs) may be at a heightened risk of benzbromarone-induced hepatotoxicity. pms 92-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 23932726-2 2014 The uricosuric drug benzbromarone is an alternative treatment but CYP2C9 poor metabolisers (PMs) may be at a heightened risk of benzbromarone-induced hepatotoxicity. Benzbromarone 128-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 23932726-10 2014 CONCLUSION: Prospective CYP2C9 genotyping of Caucasian gout patients may be warranted for benzbromarone, whereas the low frequencies of CYP2C9 PM alleles in Polynesians suggests that the CYP2C9 polymorphism may be of little or no relevance to benzbromarone prescribing in this population. Benzbromarone 90-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24360850-1 2014 BACKGROUND: Genetic polymorphisms in the CYP2C9 and VKORC1 genes have been linked to sensitivity of the anticoagulant drug warfarin. Warfarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 23844998-0 2014 Effect of 36 CYP2C9 variants found in the Chinese population on losartan metabolism in vitro. Losartan 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 23844998-4 2014 The aim of this study was to assess the catalytic activities of 36 CYP2C9 variants found in the Chinese population toward losartan in vitro. Losartan 122-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 23844998-6 2014 Insect microsomes expressing the 36 CYP2C9 variants were incubated with 0.5-25 muM losartan for 30 min at 37 C. Next, the products were extracted, and signal detection was performed using high-performance liquid chromatography. Losartan 83-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 23844998-11 2014 These findings suggest that more attention should be paid to subjects carrying these infrequent CYP2C9 alleles when administering losartan in the clinic. Losartan 130-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 24615047-0 2014 Effect of CYP2C9*3 mutant variants on meloxicam pharmacokinetics in a healthy Chinese population. Meloxicam 38-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 24615047-1 2014 The aim of this study was to investigate the effect of the CYP2C9*3 (CYP2C9 1075 A>C) polymorphism on meloxicam pharmacokinetics in a Chinese population. Meloxicam 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 59-65 24615047-1 2014 The aim of this study was to investigate the effect of the CYP2C9*3 (CYP2C9 1075 A>C) polymorphism on meloxicam pharmacokinetics in a Chinese population. Meloxicam 105-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 24615047-8 2014 CYP2C9*3 was found to play an important role in the metabolism of meloxicam by reducing its enzymatic activity. Meloxicam 66-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24498291-6 2014 The calculated binding equilibrium of APAP within the compact active site of CYP2A6 is able to predict the experimentally documented product ratios and is also applied to explain APAP regioselectivity in CYP1A2 and CYP2C9. Acetaminophen 38-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 215-221 24166101-3 2014 Here, we describe a bioreactor that is made by immobilizing cytochrome P450 2C9 (CYP2C9) to a poly(methyl methacrylate) surface and, as an alternative to traditional chemical synthesis, can be used to biosynthesize P450 metabolites in a plug flow bioreactor. Polymethyl Methacrylate 94-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-79 24498291-4 2014 In CYP2C9 and CYP3A4, weak interactions in an overall large active site cavity result in a fairly small binding free energy difference between APAP reactive binding states, consistent with experimental results that show little preference for resulting metabolites. Acetaminophen 143-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 3-9 24166101-3 2014 Here, we describe a bioreactor that is made by immobilizing cytochrome P450 2C9 (CYP2C9) to a poly(methyl methacrylate) surface and, as an alternative to traditional chemical synthesis, can be used to biosynthesize P450 metabolites in a plug flow bioreactor. Polymethyl Methacrylate 94-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 24753822-2 2014 In the present study, the inhibitory effects of polyozellin and thelephoric acid on 9 cytochrome P450 (CYP) family members (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4) were examined in pooled human liver microsomes (HLMs) using a cocktail probe assay. polyozellin 48-59 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 156-162 24166101-6 2014 Our results show that the immobilization of CYP2C9 enzymes to a PMMA surface represents a viable and alternative approach to the preparation of CYP2C9 metabolites for toxicity testing. Polymethyl Methacrylate 64-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 24166101-6 2014 Our results show that the immobilization of CYP2C9 enzymes to a PMMA surface represents a viable and alternative approach to the preparation of CYP2C9 metabolites for toxicity testing. Polymethyl Methacrylate 64-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 24186263-10 2014 RESULTS: In vitro AZD2066 inhibited CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP2D6. AZD2066 18-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 24160757-5 2014 Clinical pharmacogenetic data further support CYP2C9 as a significant contributor to THC metabolism, and a pharmacokinetic interaction study using ketoconazole with oromucosal cannabis extract further supports CYP3A4 as a significant metabolic pathway for THC and CBD. Dronabinol 85-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 24322170-0 2014 Strongly increased exposure of meloxicam in CYP2C9*3/*3 individuals. Meloxicam 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 24287930-0 2014 Variability of warfarin dose response associated with CYP2C9 and VKORC1 gene polymorphisms in Chinese patients. Warfarin 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 24038489-8 2014 The CYP2C9 pathway (4-hydroxyflurbiprofen formation clearance) was similar in all the patients with CKD and was concordant with previous reports of patients with end-stage renal disease (ESRD) and healthy controls. 4'-hydroxyflurbiprofen 20-41 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 24322170-4 2014 RESULTS: A nine-fold lower apparent oral clearance and an eight-fold higher AUC0- of single-dose meloxicam were observed in CYP2C9*3/*3 individuals when compared with CYP2C9*1/*1 individuals. Meloxicam 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 24322170-5 2014 CYP2C9*3/*3 individuals also showed markedly increased inhibition of thromboxane B2 generation by meloxicam. Thromboxane B2 69-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24322170-5 2014 CYP2C9*3/*3 individuals also showed markedly increased inhibition of thromboxane B2 generation by meloxicam. Meloxicam 98-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24533715-1 2014 Clinical factors, demographic variables and variations in two genes, CYP2C9 and VKORC1, have been shown to contribute to the variability in warfarin dose requirements among adult patients. Warfarin 140-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 24322170-6 2014 CONCLUSION: These results indicate that CYP2C9*3/*3 individuals may be at a higher risk for concentration-dependent adverse effects during long-term treatment with standard doses of meloxicam. Meloxicam 182-191 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 24302581-3 2014 In this study, we show that targeting the formation of proangiogenic epoxyeicosatrienoic acids (EET) by the cytochrome P450 arachidonic acid epoxygenases (Cyp2c) represents a new and safe mechanism to treat NSCLC growth and progression. epoxyeicosatrienoic acids 69-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-160 24387788-0 2014 Effects of anthocyanidins and anthocyanins on the expression and catalytic activities of CYP2A6, CYP2B6, CYP2C9, and CYP3A4 in primary human hepatocytes and human liver microsomes. Anthocyanins 11-25 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 24387788-0 2014 Effects of anthocyanidins and anthocyanins on the expression and catalytic activities of CYP2A6, CYP2B6, CYP2C9, and CYP3A4 in primary human hepatocytes and human liver microsomes. Anthocyanins 30-42 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 24387788-3 2014 The current study evaluated the effects of anthocyanidins and anthocyanins on the expression and catalytic activity of major drug-metabolizing enzymes CYP2C9, CYP2A6, CYP2B6, and CYP3A4 in primary cultures of human hepatocytes and human liver microsomes. Anthocyanins 43-57 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 24387788-3 2014 The current study evaluated the effects of anthocyanidins and anthocyanins on the expression and catalytic activity of major drug-metabolizing enzymes CYP2C9, CYP2A6, CYP2B6, and CYP3A4 in primary cultures of human hepatocytes and human liver microsomes. Anthocyanins 62-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 24387788-8 2014 Catalytic activities of CYP2C9, CYP2A6, CYP2B6, and CYP3A4 enzymes were inhibited by all anthocyanidins to different extents (e.g., delphinidin inhibits CYP3A4 by >90% at 100 muM with IC50 = 32 muM). Anthocyanins 89-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24387788-8 2014 Catalytic activities of CYP2C9, CYP2A6, CYP2B6, and CYP3A4 enzymes were inhibited by all anthocyanidins to different extents (e.g., delphinidin inhibits CYP3A4 by >90% at 100 muM with IC50 = 32 muM). delphinidin 132-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24302581-3 2014 In this study, we show that targeting the formation of proangiogenic epoxyeicosatrienoic acids (EET) by the cytochrome P450 arachidonic acid epoxygenases (Cyp2c) represents a new and safe mechanism to treat NSCLC growth and progression. EET 96-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 155-160 24636047-7 2014 One of the main factors affecting the efficacy of Clopidogrel is the phenotype associated with polymorphisms in the gene CYP 2C9. Clopidogrel 50-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 121-128 26161443-0 2014 A chiral HPLC-MS/MS method for simultaneous quantification of warfarin enantiomers and its major hydroxylation metabolites of CYP2C9 and CYP3A4 in human plasma. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 126-132 26161443-2 2014 Enantiomeric separation and quantification of warfarin enantiomers and clinically important major hydroxylation metabolites are essential for drug interaction studies and phenotypic characterization of CYP2C9 and CYP3A4, the major cytochrome P450 (CYP) enzymes involved in warfarin metabolism. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 202-208 26161443-3 2014 Here, we describe the development and validation of a chiral high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS)-based quantification of R-warfarin, S-warfarin, S-7-hydroxywarfarin (the major CYP2C9 metabolite) and (9R;10S)-10-hydroxywarfarin (the CYP3A4 metabolite) in human plasma. 7-hydroxywarfarin 186-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 217-223 25318916-0 2014 Frequencies of polymorphisms in CYP2C9 and VKORC1 genes influencing warfarin metabolism in Slovak population: implication for clinical practice. Warfarin 68-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 24423593-2 2014 P-gp (fexofenadine) and CYP-specific substrates (caffeine for CYP1A2, bupropion for CYP2B6, flurbiprofen for CYP2C9, omeprazole for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4) and their metabolites were extracted from DBS (10 microl) using methanol. Flurbiprofen 92-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 109-115 25318916-1 2014 OBJECTIVES: The study was aimed at establishing an effective molecular-genetic method for detecting polymorphisms in genes CYP2C9 and VKORC1, which affect the pharmacogenetics of warfarin, and at determining their prevalence in Slovak population. Warfarin 179-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 123-129 25318916-4 2014 It was proven that genetic variability in two genes, CYP2C9 a VKORC1, has a significant influence on the individual"s response to the dosage of warfarin. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 25318916-6 2014 RESULTS: Due to the combination of frequent alleles CYP2C9*2, CYP2C9*3 and VKORC1*2 in Slovak population we determine that 25% of population need a standard 5-mg daily dose of warfarin, while 44%, 23%, and 8% need 4 mg, 3 mg and 2 mg of warfarin per day. Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 25318916-6 2014 RESULTS: Due to the combination of frequent alleles CYP2C9*2, CYP2C9*3 and VKORC1*2 in Slovak population we determine that 25% of population need a standard 5-mg daily dose of warfarin, while 44%, 23%, and 8% need 4 mg, 3 mg and 2 mg of warfarin per day. Warfarin 176-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. Flavonoids 91-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 24627811-0 2014 Warfarin Dosing in a Patient with CYP2C9(*)3(*)3 and VKORC1-1639 AA Genotypes. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 24627811-1 2014 Genetic factors most correlated with warfarin dose requirements are variations in the genes encoding the enzymes cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR). Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 113-132 24627811-1 2014 Genetic factors most correlated with warfarin dose requirements are variations in the genes encoding the enzymes cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKOR). Warfarin 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 24627811-2 2014 Patients receiving warfarin who possess one or more genetic variations in CYP2C9 and VKORC1 are at increased risk of adverse drug events and require significant dose reductions to achieve a therapeutic international normalized ratio (INR). Warfarin 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 24492587-4 2014 The amino acids in these positions have opposite charges in CYP2C9 and 2C19; the former has lysines in both positions (Lys72 and Lys241), and the latter has glutamic acids (Glu72 and Glu241). Lysine 92-99 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. Quercetin 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. Rutin 113-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. naringenin 120-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 25109798-3 2014 Extensive evidence suggests that common genetic variants in CYP2C9 and VKORC1 genes together with a number of clinical factors are important determinants of the coumarin dose variability. coumarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. naringin 135-143 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 24479689-6 2014 Moreover, an integrated in silico approach for the prediction of Phase I metabolism of the flavonoids quercetin, rutin, naringenin and naringin, which provided useful information about the most likely metabolites of these flavonoids and their interactions with amino acid residues of CYP2C9, is described. Flavonoids 222-232 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 284-290 24695277-3 2014 Sequential metabolic reactions by Phase I and then Phase II enzymes were found in diclofenac [CYP2C9 and UDP-glucuronyltransferases (UGTs)], midazolam (CYP3A4 and UGTs) and propranolol (CYP1A2/2D6 and UGTs). Diclofenac 82-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 25429674-5 2014 In in vivo pharmacokinetic studies, itraconazole was found to be a potent clinically important inhibitor of CYP3A4/5 substrates, and fluconazole and voriconazole increased the blood/plasma concentrations of not only CYP3A4/5 substrates but also CYP2C9 substrates. Itraconazole 36-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 245-251 25429674-5 2014 In in vivo pharmacokinetic studies, itraconazole was found to be a potent clinically important inhibitor of CYP3A4/5 substrates, and fluconazole and voriconazole increased the blood/plasma concentrations of not only CYP3A4/5 substrates but also CYP2C9 substrates. Fluconazole 133-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 245-251 25429674-5 2014 In in vivo pharmacokinetic studies, itraconazole was found to be a potent clinically important inhibitor of CYP3A4/5 substrates, and fluconazole and voriconazole increased the blood/plasma concentrations of not only CYP3A4/5 substrates but also CYP2C9 substrates. Voriconazole 149-161 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 245-251 25429674-6 2014 Miconazole was a potent inhibitor of all P450s investigated in vitro, although there are few detailed studies on the clinical significance of this except for CYP2C9. Miconazole 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 24695277-3 2014 Sequential metabolic reactions by Phase I and then Phase II enzymes were found in diclofenac [CYP2C9 and UDP-glucuronyltransferases (UGTs)], midazolam (CYP3A4 and UGTs) and propranolol (CYP1A2/2D6 and UGTs). Propranolol 173-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 24825094-1 2014 CYP2C9 metabolizes approximately 20% of clinically used drugs, including the narrow therapeutic window drugs warfarin and phenytoin. Warfarin 109-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24825094-1 2014 CYP2C9 metabolizes approximately 20% of clinically used drugs, including the narrow therapeutic window drugs warfarin and phenytoin. Phenytoin 122-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24019055-8 2014 CONCLUSION: Genetic polymorphisms of CYP2C9, VKORC1, CYP4F2 and GGCX are important predictive factors of warfarin maintenance dose, and the developed algorithm will be useful to predict the required maintenance and/or starting warfarin dose in South Indian populations. Warfarin 105-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 24019055-0 2014 Effect of CYP2C9, VKORC1, CYP4F2 and GGCX genetic variants on warfarin maintenance dose and explicating a new pharmacogenetic algorithm in South Indian population. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 24019055-5 2014 Patients with the CYP2C9*1/*2, *1/*3 and *2/*3 variant genotypes required a 51.0 (2.8 mg), 60.9 (2.3 mg) and 62.2 % (2.2 mg) lower daily maintenance dose of warfarin, respectively, than those patients with the CYP2C9*1/*1 wild-type genotype (5.2 mg) (p<0.0001). Warfarin 157-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 24019055-8 2014 CONCLUSION: Genetic polymorphisms of CYP2C9, VKORC1, CYP4F2 and GGCX are important predictive factors of warfarin maintenance dose, and the developed algorithm will be useful to predict the required maintenance and/or starting warfarin dose in South Indian populations. Warfarin 227-235 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 37-43 24019055-6 2014 The genetic variants of CYP2C9, VKORC1 and GGCX were associated with decreased warfarin dose, except for rs7196161, rs7294 and rs2108622 which were associated with an increased warfarin dose. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24899908-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes dehydroepiandrosterone-sulfate (DHEA-S), but in elderly people the amount of DHEA-S remaining after CYP2C9 metabolization may be insufficient for optimal health. Dehydroepiandrosterone Sulfate 41-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24899908-1 2014 Cytochrome P450 2C9 (CYP2C9) metabolizes dehydroepiandrosterone-sulfate (DHEA-S), but in elderly people the amount of DHEA-S remaining after CYP2C9 metabolization may be insufficient for optimal health. Dehydroepiandrosterone Sulfate 41-71 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 24899908-4 2014 The interaction between 4-methoxymagndialdehyde and CYP2C9 is more intense than with other TCM compounds, but the simulation is longer. 4'-methoxymagndialdehyde 24-47 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 25061471-11 2014 Fisetin showed potent inhibitory effects on CYP2C9, CYP2C19, and CYP1A2. fisetin 0-7 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 24464600-0 2014 CYP2C9*2 allele increases risk for hypoglycemia in POR*1/*1 type 2 diabetic patients treated with sulfonylureas. Sulfonylurea Compounds 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24464600-10 2014 Therefore, POR*28 allele is an important source of CYP2C9 activity variability and combined with CYP2C9 gene poly-morphisms may explain individual variability in the effect of sulfonylureas. Sulfonylurea Compounds 176-189 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 24799741-0 2014 Investigating the Role of Plasma Glucose Concentration as a Phenotypic Marker for CYP2C9 Genetic Variants, in the Diabetic Population of Gujarat. Glucose 33-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 24799741-7 2014 Patients with CYP2C9*1/*3 genotype showed greater mean glucose drop per milligram of drug values than patients with CYP2C9*1/*1 wild-type genotype for both glipizide and glimepiride while patients with CYP2C9*2/*3 genotype showed greater drop than patients with CYP2C9*1/*1 genotype only in the glipizide-treatment group. Glucose 55-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 24799741-7 2014 Patients with CYP2C9*1/*3 genotype showed greater mean glucose drop per milligram of drug values than patients with CYP2C9*1/*1 wild-type genotype for both glipizide and glimepiride while patients with CYP2C9*2/*3 genotype showed greater drop than patients with CYP2C9*1/*1 genotype only in the glipizide-treatment group. Glipizide 156-165 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 24799741-7 2014 Patients with CYP2C9*1/*3 genotype showed greater mean glucose drop per milligram of drug values than patients with CYP2C9*1/*1 wild-type genotype for both glipizide and glimepiride while patients with CYP2C9*2/*3 genotype showed greater drop than patients with CYP2C9*1/*1 genotype only in the glipizide-treatment group. glimepiride 170-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 24799741-7 2014 Patients with CYP2C9*1/*3 genotype showed greater mean glucose drop per milligram of drug values than patients with CYP2C9*1/*1 wild-type genotype for both glipizide and glimepiride while patients with CYP2C9*2/*3 genotype showed greater drop than patients with CYP2C9*1/*1 genotype only in the glipizide-treatment group. Glipizide 295-304 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 24799741-8 2014 The presence of CYP2C9*3 allele significantly affected plasma glucose drop per milligram of drug values in patients taking glipizide and glimepiride, while effects of CYP2C9*2 allele were insignificant. Glucose 62-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 24799741-8 2014 The presence of CYP2C9*3 allele significantly affected plasma glucose drop per milligram of drug values in patients taking glipizide and glimepiride, while effects of CYP2C9*2 allele were insignificant. Glipizide 123-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 24799741-8 2014 The presence of CYP2C9*3 allele significantly affected plasma glucose drop per milligram of drug values in patients taking glipizide and glimepiride, while effects of CYP2C9*2 allele were insignificant. glimepiride 137-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 24799741-9 2014 Further studies are needed to confirm the effects of CYP2C9*2 allele on plasma glucose drop per milligram of drug values. Glucose 79-86 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 53-59 25075423-0 2014 Characterization of a novel CYP2C9 mutation (1009C>A) detected in a warfarin-sensitive patient. Warfarin 71-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 28-34 25075423-2 2014 Recent studies have shown that polymorphic alleles within the CYP2C9, VKORC1, and CYP4F2 genes are related to the warfarin dosage requirement. Warfarin 114-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 25075423-3 2014 In this study, a novel non-synonymous mutation (1009C>A) in CYP2C9 was detected in a warfarin-hypersensitive patient, while the other two candidate genes were both found to be homozygous for the wild-type alleles. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Tolbutamide 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Tolbutamide 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Tolbutamide 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Tolbutamide 111-122 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Losartan 127-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 197-203 25075423-5 2014 When expressed in insect cell microsomes, the relative intrinsic clearance values of the CYP2C9.58 variant for tolbutamide and losartan were quite similar to those of the typical defective variant CYP2C9.3, whereas the clearance value of CYP2C9.58 for diclofenac was slightly higher than that of another typical defective variant CYP2C9.2. Diclofenac 252-262 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 25343299-7 2014 Only thujopsene was found to be a mechanism-based inhibitor of CYP2C8, CYP2C9, and CYP2C19. thujopsene 5-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 25343299-8 2014 Cedrol and thujopsene weakly inhibited CYP2C8, CYP2C9, and CYP2C19 activities, but beta-cedrene did not. cedrol 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 25343299-8 2014 Cedrol and thujopsene weakly inhibited CYP2C8, CYP2C9, and CYP2C19 activities, but beta-cedrene did not. thujopsene 11-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 25335408-1 2014 The goal of this work was to study cytochrome-450 (CYP) 2C9 (CYP2C9) gene polymorphism in patients with tuberculosis (TB) and its meaning for development, progress, and outcome of TB, for the pharmacokinetics of the antituberculosis antibiotic rifampicin on the basis of the southern region of Ukraine. Rifampin 244-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 35-59 25335408-1 2014 The goal of this work was to study cytochrome-450 (CYP) 2C9 (CYP2C9) gene polymorphism in patients with tuberculosis (TB) and its meaning for development, progress, and outcome of TB, for the pharmacokinetics of the antituberculosis antibiotic rifampicin on the basis of the southern region of Ukraine. Rifampin 244-254 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 24245489-5 2014 Genotyping for various CYP single-nucleotide polymorphisms showed that the patient carried mutant alleles encoding enzymes CYP2B6 and CYP2C9 involved in prasugrel metabolic pathway. Prasugrel Hydrochloride 153-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 134-140 25185986-5 2014 In this review, we discuss the implications of polymorphisms in the cytochrome P-450 enzyme 2C9 (CYP2C9) and Vitamin K Epoxide Reductase Enzyme Complex subunit 1 (VKORC1) as they relate to therapeutic warfarin dosing. Warfarin 201-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-95 25185986-5 2014 In this review, we discuss the implications of polymorphisms in the cytochrome P-450 enzyme 2C9 (CYP2C9) and Vitamin K Epoxide Reductase Enzyme Complex subunit 1 (VKORC1) as they relate to therapeutic warfarin dosing. Warfarin 201-209 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 97-103 25492821-4 2014 VKORC1 and CYP2C9 are two genes responsible for warfarin metabolism. Warfarin 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24464600-1 2014 It is previously shown that carriers of the defective allele CYP2C9*3 that leads to impaired sulfonylurea metabolism are at increased sulfonylurea-induced hypoglycemia risk due to diminished drug metabolism, whereas no effect of CYP2C9*2 allele was found. Sulfonylurea Compounds 93-105 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 61-67 24464600-8 2014 In a model adjusted for age, BMI, duration of T2DM and renal function, and POR*1/*1 entered as a selection variable, CYP2C9*2 allele increased the hypoglycemia risk in response to sulfonylurea (odds ratio: 3.218, p=0.031). Sulfonylurea Compounds 180-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 24464600-9 2014 In conclusion, our results suggest that POR*28 allele is masking the association of CYP2C9*2 allele with sulfonyl-urea-induced hypoglycemia. Sulfonylurea Compounds 105-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 23863877-0 2013 Association of CYP2C9*2 with bosentan-induced liver injury. Bosentan 29-37 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 15-21 24043253-3 2013 We tested the hypothesis that regional inhibition of endothelium-derived hyperpolarizing factor with the cytochrome P-450 2C9 antagonist fluconazole, alone or combined with the NO synthase antagonist N(G)-monomethyl-L-arginine (L-NMMA), attenuates hypoxia-induced vasodilation. Fluconazole 137-148 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-125 23863877-6 2013 Bosentan metabolism by CYP2C9*2 in vitro was significantly reduced compared with CYP2C9*1 and was comparable to that by CYP2C9*3. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 23863877-7 2013 These results suggest that CYP2C9*2 is a potential genetic marker for prediction of bosentan-induced liver injury and warrants investigation for the optimization of bosentan treatment. Bosentan 84-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 23863877-7 2013 These results suggest that CYP2C9*2 is a potential genetic marker for prediction of bosentan-induced liver injury and warrants investigation for the optimization of bosentan treatment. Bosentan 165-173 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 23863877-6 2013 Bosentan metabolism by CYP2C9*2 in vitro was significantly reduced compared with CYP2C9*1 and was comparable to that by CYP2C9*3. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 23863877-6 2013 Bosentan metabolism by CYP2C9*2 in vitro was significantly reduced compared with CYP2C9*1 and was comparable to that by CYP2C9*3. Bosentan 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 24046330-6 2013 Spectral scanning of the reaction between CYP2C9 and noscapine revealed the formation of an absorption spectrum at 458 nm, indicating the formation of a metabolite-intermediate complex. Noscapine 53-62 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 23974699-7 2013 CYP1A2 and CYP2D6 enzyme activities showed no significant changes, and CYP2C9 enzyme activity was increased with rifampicin administration, with a tendency toward statistical significance. Rifampin 113-123 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 24046330-4 2013 Our enzyme kinetics data obtained from human liver microsomes and recombinant CYP2C9 proteins indicate that noscapine is a competitive inhibitor of the (S)-warfarin 7-hydroxylation reaction by CYP2C9. Noscapine 108-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 24046330-7 2013 Surprisingly, noscapine is a 2- to 3-fold more efficient inactivator of CYP2C9.2 and CYP2C9.3 variants than it is of the wild type, by unknown mechanisms. Noscapine 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24046330-4 2013 Our enzyme kinetics data obtained from human liver microsomes and recombinant CYP2C9 proteins indicate that noscapine is a competitive inhibitor of the (S)-warfarin 7-hydroxylation reaction by CYP2C9. Noscapine 108-117 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 24046330-4 2013 Our enzyme kinetics data obtained from human liver microsomes and recombinant CYP2C9 proteins indicate that noscapine is a competitive inhibitor of the (S)-warfarin 7-hydroxylation reaction by CYP2C9. Warfarin 152-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 78-84 24046330-7 2013 Surprisingly, noscapine is a 2- to 3-fold more efficient inactivator of CYP2C9.2 and CYP2C9.3 variants than it is of the wild type, by unknown mechanisms. Noscapine 14-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-91 24046330-4 2013 Our enzyme kinetics data obtained from human liver microsomes and recombinant CYP2C9 proteins indicate that noscapine is a competitive inhibitor of the (S)-warfarin 7-hydroxylation reaction by CYP2C9. Warfarin 152-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 24046330-8 2013 Based on the inhibitory kinetic data, (S)-warfarin exposure is predicted to increase up to 7-fold (depending on CYP2C9 genotypes) upon noscapine coadministration, mainly due to mechanism-based inactivation of CYP2C9 by noscapine. Warfarin 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 24046330-8 2013 Based on the inhibitory kinetic data, (S)-warfarin exposure is predicted to increase up to 7-fold (depending on CYP2C9 genotypes) upon noscapine coadministration, mainly due to mechanism-based inactivation of CYP2C9 by noscapine. Warfarin 38-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 24046330-8 2013 Based on the inhibitory kinetic data, (S)-warfarin exposure is predicted to increase up to 7-fold (depending on CYP2C9 genotypes) upon noscapine coadministration, mainly due to mechanism-based inactivation of CYP2C9 by noscapine. Noscapine 135-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 24046330-8 2013 Based on the inhibitory kinetic data, (S)-warfarin exposure is predicted to increase up to 7-fold (depending on CYP2C9 genotypes) upon noscapine coadministration, mainly due to mechanism-based inactivation of CYP2C9 by noscapine. Noscapine 135-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 209-215 24046330-9 2013 Together, these results indicate that mechanism-based inhibition of CYP2C9 by noscapine may dramatically alter pharmacokinetics of warfarin and provide a basis for warfarin dosage adjustment when noscapine is coadministered. Noscapine 78-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 24046330-9 2013 Together, these results indicate that mechanism-based inhibition of CYP2C9 by noscapine may dramatically alter pharmacokinetics of warfarin and provide a basis for warfarin dosage adjustment when noscapine is coadministered. Warfarin 131-139 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 24046330-9 2013 Together, these results indicate that mechanism-based inhibition of CYP2C9 by noscapine may dramatically alter pharmacokinetics of warfarin and provide a basis for warfarin dosage adjustment when noscapine is coadministered. Warfarin 164-172 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 24046330-9 2013 Together, these results indicate that mechanism-based inhibition of CYP2C9 by noscapine may dramatically alter pharmacokinetics of warfarin and provide a basis for warfarin dosage adjustment when noscapine is coadministered. Noscapine 196-205 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 68-74 23941071-7 2013 CONCLUSION: These findings indicate that individuals carrying the CYP2C19 rs3814637CC or CYP2C9 rs1057910AA or GGCX rs699664AA genotype needed higher warfarin doses in the Chinese population. Warfarin 150-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 23996211-0 2013 Evaluation of the relationship between polymorphisms in CYP2C8 and CYP2C9 and the pharmacokinetics of celecoxib. Celecoxib 102-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 23996211-1 2013 Celecoxib is metabolized by enzymes of the cytochrome P450 (CYP450) superfamily, mainly CYP2C9 and CYP3A4. Celecoxib 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 23996211-2 2013 Polymorphisms in the CYP2C9 gene have been associated with decreased enzyme activity and alteration of celecoxib pharmacokinetic parameters. Celecoxib 103-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 23996211-8 2013 In conclusion, the recommended dose of celecoxib should be decreased in CYP2C9*3 carriers, especially in homozygous subjects. Celecoxib 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 25505570-9 2013 Additional studies were done to ascertain if ajulemic acid can inhibit the activities of five principal human cytochrome P450 isozymes; CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4/5. lenabasum 45-58 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 144-150 23839801-10 2013 SYBR Green-based multiplex allele-specific real-time PCR was used for genotyping of CYP2C9 (1075A>C) and VKORC1 (1173C>T) polymorphisms. sybr green 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 23839801-11 2013 Linear regression analysis, including the variables age, gender, CYP2C9 and VKORC1 SNP genotypes, was run to derive the best model for estimating the warfarin dose that achieves an INR of 2-3. Warfarin 150-158 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 23839801-14 2013 RESULTS: In the study cohort, warfarin dose/week in VKORC1 TT subjects was statistically significantly lower than in VKORC1 CC/CT subjects (p=0.032), while there was no statistically significant difference in warfarin dose/week between CYP2C9*1*1 and *1*3 (p=0.925). Warfarin 30-38 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 236-242 24503429-0 2013 [A randomized controlled study of the VKORC1 and CYP2C9 genotypes in guiding warfarin therapy for pulmonary thromboembolism]. Warfarin 77-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 24521898-2 2013 METHODS: Genomic DNAs were extracted from the warfarin hypersensitive patient and used for genetic screening of CYP2C9, VKORC1 and CYP4F2 by direct sequencing. Warfarin 46-54 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 112-118 23679834-0 2013 Monkey liver cytochrome P450 2C9 is involved in caffeine 7-N-demethylation to form theophylline. Caffeine 48-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-32 23679834-0 2013 Monkey liver cytochrome P450 2C9 is involved in caffeine 7-N-demethylation to form theophylline. Theophylline 83-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-32 24521898-11 2013 When expressed in insect cell microsomes, the relative clearance values of CYP2C9 *2, *3, *13 and L467P variants to tolbutamide were 91.58%, 13.55%, 0.11% and 1.59% to that of wild-type protein respectively. Tolbutamide 116-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 75-81 23807523-0 2013 Effects of amiodarone, thyroid hormones and CYP2C9 and VKORC1 polymorphisms on warfarin metabolism: a review of the literature. Warfarin 79-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 44-50 23872824-8 2013 CONCLUSIONS: Substantial inhibition of CYP3A- and modest induction of CYP2C9- activity were observed with danoprevir/r and low-dose ritonavir. danoprevir 106-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 23872824-8 2013 CONCLUSIONS: Substantial inhibition of CYP3A- and modest induction of CYP2C9- activity were observed with danoprevir/r and low-dose ritonavir. r 89-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 23872824-8 2013 CONCLUSIONS: Substantial inhibition of CYP3A- and modest induction of CYP2C9- activity were observed with danoprevir/r and low-dose ritonavir. Ritonavir 132-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 23649958-1 2013 Warfarin, an anticoagulant with a narrow therapeutic window, is largely metabolized by cytochrome P450 2C9. Warfarin 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 87-106 24071466-8 2013 Polymorphism of CYP2C9 and VKORC1 were related to the dosage of warfarin but not determinant of TTR. Warfarin 64-72 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 24380239-5 2013 Seven subjects (2.97%) were poor metabolites (PMs) for CYP2C9 because of the *2/*2 and *3/*3 genotype. pms 46-49 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 55-61 24222221-0 2013 Genetic contribution of CYP2C9, CYP2C19, and APOE variants in acenocoumarol response. Acenocoumarol 62-75 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 24222221-4 2013 The objective of this study was to determine the contribution of CYP2C9, CYP2C19, and APOE polymorphisms to the variations in response to the doses of acenocoumarol, which is the main anticoagulant prescribed to the Mexican population. Acenocoumarol 151-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 23852652-5 2013 Roles for CYP3A4, CYP2C9, CYP2C19 and CYP2B6 in the metabolism of ospemifene and its two main metabolites, 4--hydroxyospemifene and 4"-hydroxyospemifene, were confirmed. Ospemifene 66-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 23932037-0 2013 Influence of CYP2C9 and VKORC1 genotypes on the risk of hemorrhagic complications in warfarin-treated patients: a systematic review and meta-analysis. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 23932037-14 2013 CONCLUSIONS: Both CYP2C9 and VKORC1 genotypes are associated with an increased risk for warfarin over-anticoagulation, with VKORC1 c. -1639G > A more sensitive early in the course of anticoagulation. Warfarin 88-96 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 23932037-15 2013 CYP2C9*3 is the main genetic risk factor for warfarin hemorrhagic complications. Warfarin 45-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23852652-8 2013 Strong CYP3A or CYP2C9 inducers (e.g. rifampicin) would be expected to decrease the exposure to ospemifene. Rifampin 38-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 23852652-8 2013 Strong CYP3A or CYP2C9 inducers (e.g. rifampicin) would be expected to decrease the exposure to ospemifene. Ospemifene 96-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 16-22 24174816-2 2013 AIMS: This study was conducted to determine the inhibitory effects of mitragynine on cytochrome P450 2C9, 2D6 and 3A4 activities. mitragynine 70-81 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 85-104 24174816-6 2013 RESULTS: Assessment using recombinant enzymes showed that mitragynine gave the strongest inhibitory effect on CYP2D6 with an IC50 value of 0.45+-0.33 mM, followed by CYP2C9 and CYP3A4 with IC50 values of 9.70+-4.80 and 41.32+-6.74 muM respectively. mitragynine 58-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 166-172 24174816-7 2013 Positive inhibitors appropriate for CYP2C9, CYP2D6, and CYP3A4 which are sulfaphenazole, quinidine and ketoconazole were used respectively. Sulfaphenazole 73-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 24174816-7 2013 Positive inhibitors appropriate for CYP2C9, CYP2D6, and CYP3A4 which are sulfaphenazole, quinidine and ketoconazole were used respectively. Quinidine 89-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 24174816-7 2013 Positive inhibitors appropriate for CYP2C9, CYP2D6, and CYP3A4 which are sulfaphenazole, quinidine and ketoconazole were used respectively. Ketoconazole 103-115 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 24174816-10 2013 Mitragynine noncompetitively inhibits CYP2C9 and CYP2D6 activities with the Ki values of 61.48 and 12.86 muM respectively. mitragynine 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 23688605-0 2013 First report of warfarin dose requirements in patients possessing the CYP2C9*12 allele. Warfarin 16-24 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 70-76 23688605-3 2013 Cytochrome P450 (CYP) 2C9 is the enzyme primarily responsible for the metabolism of S-warfarin. Warfarin 84-94 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-25 23688605-12 2013 This is the first report of patients with the rare CYP2C9*12 genotype and lower warfarin dose requirements. Warfarin 80-88 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 51-57 24005963-5 2013 These in vitro results indicate that honokiol has the potential to cause pharmacokinetic drug interactions with other co-administered drugs metabolized by CYP1A2, CYP2C8, CYP2C9, CYP2C19, and UGT1A9. honokiol 37-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 171-177 24023924-0 2013 Distal effect of amino acid substitutions in CYP2C9 polymorphic variants causes differences in interatomic interactions against (S)-warfarin. Warfarin 128-140 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 24023924-5 2013 To relate distal effects of these mutations and defective drug metabolisms, simulations of CYP2C9 binding to anti-coagulant (S)-warfarin were performed as a system model. Sulfur 124-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 24023924-5 2013 To relate distal effects of these mutations and defective drug metabolisms, simulations of CYP2C9 binding to anti-coagulant (S)-warfarin were performed as a system model. Warfarin 128-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 91-97 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. coumarin 139-147 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23770984-5 2013 RESULTS: All three gingerols potently inhibited CYP2C9 activity, exerted moderate inhibition on CYP2C19 and CYP3A4, and weak inhibion on CYP2D6. gingerol 19-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 48-54 23770984-6 2013 8-Gingerol was the most potent in inhibition of P450 enzymes with IC50 values of 6.8, 12.5, 8.7, and 42.7 mumol/L for CYP2C9, CYP2C19, CYP3A4, and CYP2D6, respectively. 8-gingerol 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-124 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. Propranolol 149-160 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. aniline 162-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. Chlorzoxazone 171-184 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. 4-nitrophenol 186-199 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23287855-4 2013 In vitro analysis suggested that the purified CYP102A5 metabolizes typical substrates of human CYP2C9, CYP2D6, CYP2E1, and CYP3A4, such as coumarin, propranolol, aniline, chlorzoxazone, p-nitrophenol, and nifedipine. Nifedipine 205-215 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23504614-6 2013 SPZ also prevented CA-induced mitochondrial membrane depolarization and ECs death, implicating CYP2C9 in mediating the cellular response upon CA treatment. Sulfaphenazole 0-3 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23436703-2 2013 However, multiple genes are likely to contribute, either acting independently or epistatically, with the CYP2C9-VKORC1-warfarin test panel, an example of a clinically used gene-gene-dug interaction. Warfarin 119-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 105-111 23726967-1 2013 PURPOSE: The aim of the present study was to investigate the genetic variability of VKORC1, CYP2C9 and CYP4F2 genes in patients who required a very low and high warfarin dose, in order to identify novel variants that could help to explain the particular extreme dose requirements. Warfarin 161-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 92-98 23682017-4 2013 In this review, we focus on the important CYP2C9 and VKORC1 variants involved in warfarin response, our current understanding of warfarin disposition and pharmacogenomics, and recent warfarin pharmacogenetic studies in pediatric patients. Warfarin 81-89 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 42-48 24024895-0 2013 CYP2C9 and ABCG2 polymorphisms as risk factors for developing adverse drug reactions in renal transplant patients taking fluvastatin: a case-control study. Fluvastatin 121-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 24024895-1 2013 AIM: To investigate whether an association exists between fluvastatin-induced adverse drug reactions (ADRs) and polymorphisms in genes encoding the metabolizing enzyme CYP2C9 and the drug transporter ABCG2 in renal transplant recipients (RTRs). Fluvastatin 58-69 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 168-174 24024895-8 2013 CONCLUSION: Our preliminary data demonstrate an association between fluvastatin-induced ADRs in RTRs and genetic variants in the CYP2C9 and ABCG2 genes. Fluvastatin 68-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 23755828-1 2013 BACKGROUND: VKORC1 and CYP2C9 are important contributors to warfarin dose variability, but explain less variability for individuals of African descent than for those of European or Asian descent. Warfarin 60-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 23755828-14 2013 INTERPRETATION: A novel CYP2C single nucleotide polymorphism exerts a clinically relevant effect on warfarin dose in African Americans, independent of CYP2C9*2 and CYP2C9*3. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 151-157 23755828-14 2013 INTERPRETATION: A novel CYP2C single nucleotide polymorphism exerts a clinically relevant effect on warfarin dose in African Americans, independent of CYP2C9*2 and CYP2C9*3. Warfarin 100-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 23441978-1 2013 OBJECTIVES: Etravirine is a substrate and inducer of cytochrome P450 (CYP) 3A and a substrate and inhibitor of CYP2C9 and CYPC2C19. etravirine 12-22 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 23441978-3 2013 Artemether and lumefantrine are primarily metabolized by CYP3A; artemether is also metabolized to a lesser extent by CYP2B6, CYP2C9 and CYP2C19. Artemether 64-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 125-131 23930675-0 2013 Effects of polymorphisms in ABCG2, SLCO1B1, SLC10A1 and CYP2C9/19 on plasma concentrations of rosuvastatin and lipid response in Chinese patients. Rosuvastatin Calcium 94-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 56-62 23545481-4 2013 Estimates of CLint for the production of mono(2-ethyl-5-hydroxyhexyl) phthalate and mono(2-ethyl-5-oxohexyl) phthalate by human CYP2C9 1 were 4.2- and 2.6-fold greater than those by rat CYP2C6, respectively. mono(2-ethyl-5-hydroxyhexyl) phthalate 41-79 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 23545481-4 2013 Estimates of CLint for the production of mono(2-ethyl-5-hydroxyhexyl) phthalate and mono(2-ethyl-5-oxohexyl) phthalate by human CYP2C9 1 were 4.2- and 2.6-fold greater than those by rat CYP2C6, respectively. mono(2-ethyl-5-oxohexyl)phthalate 84-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 23727057-5 2013 A microdose of (14)C-tolbutamide (100 mug) was administered orally to healthy volunteers with the CYP2C9(*)1/(*)1 or CYP2C9(*)1/(*)3 diplotype. Tolbutamide 21-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 117-123 23688499-0 2013 Pregnane X receptor dependent up-regulation of CYP2C9 and CYP3A4 in tumor cells by antitumor acridine agents, C-1748 and C-1305, selectively diminished under hypoxia. Acridines 93-101 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 23688499-0 2013 Pregnane X receptor dependent up-regulation of CYP2C9 and CYP3A4 in tumor cells by antitumor acridine agents, C-1748 and C-1305, selectively diminished under hypoxia. 9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine 110-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 23688499-0 2013 Pregnane X receptor dependent up-regulation of CYP2C9 and CYP3A4 in tumor cells by antitumor acridine agents, C-1748 and C-1305, selectively diminished under hypoxia. C 1305 121-127 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 47-53 23688499-6 2013 C-1748-mediated CYP3A4 and CYP2C9 mRNA induction equal to rifampicin occurred at extremely low concentrations (0.001 and 0.01muM), whereas 10muM C-1305 induced three-times higher CYP3A4 and CYP2C9 mRNA levels than rifampicin did. 9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 23688499-6 2013 C-1748-mediated CYP3A4 and CYP2C9 mRNA induction equal to rifampicin occurred at extremely low concentrations (0.001 and 0.01muM), whereas 10muM C-1305 induced three-times higher CYP3A4 and CYP2C9 mRNA levels than rifampicin did. 9-(2'-hydroxyethylamino)-4-methyl-1-nitroacridine 0-6 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 190-196 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. Ospemifene 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. 4-hydroxyospemifene 36-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 23880855-4 2013 Ospemifene and its main metabolites 4-hydroxyospemifene and 4"-hydroxyospemifene weakly inhibited a number of CYPs (CYP2B6, CYP2C9, CYP2C19, CYP2C8, and CYP2D6) in vitro. 4-hydroxyospemifene 60-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 23880855-5 2013 However, only CYP2C9 activity was inhibited by 4-hydroxyospemifene at clinically relevant concentrations. 4-hydroxyospemifene 47-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 23737517-4 2013 The pharmacokinetics of the probe drug cocktail (flurbiprofen/fexofenadine) were altered, with formation clearance of flurbiprofen (CYP2C9 function) lower in our patient versus the average value in our study cohort, suggesting a reduction in activity. Flurbiprofen 118-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-138 24052784-1 2013 BACKGROUND: Warfarin dosage regimens using CYP2C9 and VKORC1 polymorphisms have been extensively studied in adults and is included in US Food and Drug Administration-approved warfarin labeling. Warfarin 12-20 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 24052784-1 2013 BACKGROUND: Warfarin dosage regimens using CYP2C9 and VKORC1 polymorphisms have been extensively studied in adults and is included in US Food and Drug Administration-approved warfarin labeling. Warfarin 175-183 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 43-49 24052784-11 2013 CONCLUSIONS: The proposed pediatric warfarin dosage scheme based on individual CYP2C9 (alleles *1,*2,*3) and VKORC1 rs9923231 (-1639 G>A) genotypes may offer improved dosage compared to current treatment strategies, especially in patients with variant CYP2C9 and VKORC1 alleles. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 24052784-11 2013 CONCLUSIONS: The proposed pediatric warfarin dosage scheme based on individual CYP2C9 (alleles *1,*2,*3) and VKORC1 rs9923231 (-1639 G>A) genotypes may offer improved dosage compared to current treatment strategies, especially in patients with variant CYP2C9 and VKORC1 alleles. Warfarin 36-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 255-261 24082701-0 2013 Cytochrome P450 2C9 gene polymorphism in phenytoin induced gingival enlargement: A case report. Phenytoin 41-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 24082701-7 2013 Individuals with poor metaboliser alleles of CYP2C9 gene were shown to have a reduced metabolism of phenytoin compared with wild-type alleles. Phenytoin 100-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 23615745-16 2013 In conclusion, CYP2B6 and CYP2C9 polymorphisms seem to be associated with prasugrel low-response. Prasugrel Hydrochloride 74-83 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 26-32 27181593-1 2016 AIMS/INTRODUCTION: The objective of the present study was to investigate the effects of CYP2C9*3 polymorphisms on the therapeutic response to gliclazide in type 2 diabetes patients. Gliclazide 142-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 27275407-2 2014 The frequency of heterozygous or homozygous genotypes of CYP2C9*2 & CYP2C9*3, which leads to the poor metabolizer (PM) genotype was significantly higher in HNSCC cases when compared to the healthy controls resulting in significantly increased risk in the cases. Adenosine Monophosphate 67-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 57-63 27275407-2 2014 The frequency of heterozygous or homozygous genotypes of CYP2C9*2 & CYP2C9*3, which leads to the poor metabolizer (PM) genotype was significantly higher in HNSCC cases when compared to the healthy controls resulting in significantly increased risk in the cases. Adenosine Monophosphate 67-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27275407-4 2014 Likewise, alcohol intake in cases with variant genotypes of CYP2C9*2 or CYP2C9*3 also significantly increased the HNSCC risk in cases when compared to non-alcohol users. Alcohols 10-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 27275407-4 2014 Likewise, alcohol intake in cases with variant genotypes of CYP2C9*2 or CYP2C9*3 also significantly increased the HNSCC risk in cases when compared to non-alcohol users. Alcohols 10-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27275407-4 2014 Likewise, alcohol intake in cases with variant genotypes of CYP2C9*2 or CYP2C9*3 also significantly increased the HNSCC risk in cases when compared to non-alcohol users. Alcohols 155-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 60-66 27275407-4 2014 Likewise, alcohol intake in cases with variant genotypes of CYP2C9*2 or CYP2C9*3 also significantly increased the HNSCC risk in cases when compared to non-alcohol users. Alcohols 155-162 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 22641027-1 2013 Pharmacogenetic studies have shown that genetic defects in drug-metabolizing enzymes encoded by CYP2C9, CYP2C19 genes and by the transporter ABCB1 gene can influence phenytoin (PTH) plasma levels and toxicity. Phenytoin 166-175 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 23587916-5 2013 The CYP2C9*2 allele frequencies in the Mestizo, Nahua and Teenek populations were 0.051, 0.007 and 0.005, respectively. nahua 48-53 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 4-10 23515956-5 2013 Based on in vitro enzyme kinetic parameters for S-warfarin metabolism, demographic data for Han Chinese and some scaling factors, the S-warfarin clearance (CL) was predicted for patients in the cohort with different CYP2C9 genotypes using IVIVE. Warfarin 136-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 216-222 23515956-7 2013 The warfarin maintenance dose was then predicted based on the demographic data and genotypes of CYP2C9 and VKORC1 for each patient and using the observed steady-state INR (INRss) as a target value. Warfarin 4-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 23515956-10 2013 There were significant differences in the mean warfarin daily dose in patients with different CYP2C9 and VKORC1 genotypes. Warfarin 47-55 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 94-100 23515956-11 2013 Using IVIVE, the predicted mean CL of S-warfarin for patients with CYP2C9*1/*3 (0.092 l/h, n = 11) was 57 % less than for those with wild-type *1/*1 (0.215 l/h, n = 186). Warfarin 40-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 67-73 23515956-15 2013 The mean predicted/observed warfarin doses (mg/day) for different combinations of CYP2C9 and VKORC1 genotypes were 1.54/3.75 (n = 1) for *1/*1 and GG, 3.33/3.66 (n = 36) for *1/*1 and AG, 2.31/2.41 (n = 136) for *1/*1 and AA, and 1.56/1.69 (n = 10) for *1/*3 and AA, respectively. Warfarin 28-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 82-88 23515956-16 2013 Sensitivity analysis indicated BW and genetic polymorphisms of CYP2C9 and VKORC1 were important factors affecting the warfarin maintenance dose in the study population. Warfarin 118-126 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 63-69 23515956-17 2013 CONCLUSION: The mechanistic model reported is the first to integrate IVIVE with a pharmacokinetic-pharmacodynamic model to describe the association of the warfarin maintenance dose with sex, age, BW and the genotypes of CYP2C9 and VKORC1. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 220-226 23602689-0 2013 Influence of CYP2C9 and VKORC1 gene polymorphisms on warfarin dosage, over anticoagulation and other adverse outcomes in Indian population. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 13-19 23602689-8 2013 Our results suggest that both VKORC1 and CYP2C9 genotypes showed significant impact on warfarin dose requirement, over anticoagulation in the first month of anticoagulation and number of bleeding episodes. Warfarin 87-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 22075408-3 2013 It has been shown that besides CYP2C19, CYP2C9 is involved in the hepatic biotransformation of clopidogrel to its active metabolite. Clopidogrel 95-106 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 40-46 22075408-4 2013 Consequently, CYP2C9 polymorphisms may also affect the extent of clopidogrel-mediated platelet inhibition. Clopidogrel 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 14-20 22075408-5 2013 We therefore studied the influence of CYP2C9 allelic variants on clopidogrel-mediated platelet inhibition as assessed by 5 platelet function tests. Clopidogrel 65-76 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 38-44 22075408-10 2013 CONCLUSION: Results from the VerifyNow P2Y12 assay are significantly influenced by CYP2C9 LOF variants leading to decreased clopidogrel-mediated platelet inhibition and an increased rate of HRPR. Clopidogrel 124-135 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 23579966-6 2013 Prospective randomized clinical trials are currently underway utilizing dosing algorithms that incorporate genetic polymorphisms in cytochrome P450 (CYP)2C9 and vitamin k epoxide reductase (VKORC1) to determine warfarin dosages. Warfarin 211-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 132-156 23579966-7 2013 Polymorphisms in CYP2C9 and VKORC1 account for approximately 40 % of the variance in warfarin dose. Warfarin 85-93 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 23525712-7 2013 In PE-constricted arteries, the H2S donor NaHS caused vasodilation that was inhibited by ryanodine (RyR blocker), abluminal or luminal iberiotoxin (IbTx, BKCa blocker), endothelial cell (EC) disruption, and sulfaphenazole [cytochrome P-450 2C (Cyp2C) inhibitor]. Hydrogen Sulfide 32-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-242 23525712-7 2013 In PE-constricted arteries, the H2S donor NaHS caused vasodilation that was inhibited by ryanodine (RyR blocker), abluminal or luminal iberiotoxin (IbTx, BKCa blocker), endothelial cell (EC) disruption, and sulfaphenazole [cytochrome P-450 2C (Cyp2C) inhibitor]. Hydrogen Sulfide 32-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 244-249 23525712-7 2013 In PE-constricted arteries, the H2S donor NaHS caused vasodilation that was inhibited by ryanodine (RyR blocker), abluminal or luminal iberiotoxin (IbTx, BKCa blocker), endothelial cell (EC) disruption, and sulfaphenazole [cytochrome P-450 2C (Cyp2C) inhibitor]. sodium bisulfide 42-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 223-242 23525712-7 2013 In PE-constricted arteries, the H2S donor NaHS caused vasodilation that was inhibited by ryanodine (RyR blocker), abluminal or luminal iberiotoxin (IbTx, BKCa blocker), endothelial cell (EC) disruption, and sulfaphenazole [cytochrome P-450 2C (Cyp2C) inhibitor]. sodium bisulfide 42-46 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 244-249 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Clomiphene 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Tamoxifen 12-21 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 23604525-6 2013 Clomiphene, tamoxifen and toremifene were biotransformed to 22, 23 and 18 metabolites respectively, these phase I reactions being catalyzed mainly by CYP3A4 and CYP2D6 isoforms and, to a lesser degree, by CYP3A5, CYP2B6, CYP2C9, CYP2C19 isoforms. Toremifene 26-36 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 221-227 23509226-13 2013 Apatinib was metabolized primarily by CYP3A4/5 and, to a lesser extent, by CYP2D6, CYP2C9, and CYP2E1. apatinib 0-8 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 83-89 23539296-6 2013 In addition to CYP2B6, anisomycin cotreatment potentiated an increase in CYP2A7 and CYP2C9 mRNAs but not CYP3A4 or UDP-glucuronosyltransferase 1A1 mRNAs. Anisomycin 23-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-90 23500771-4 2013 The results showed that SAG was a potent noncompetitive inhibitor of CYP2C8 activity (Ki=8.9 muM), and competitive inhibitor of CYP1A2, CYP2C9 and CYP3A4 activities (Ki=2.7, 3.8 and 2.0 muM, respectively). sanguinarine 24-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 136-142 23500771-7 2013 In vitro-in vivo extrapolation (IV-IVE) from HLM data showed that more than 35.9% of CYP1A2, CYP2C9, CYP2C8 and CYP3A4 activities in vivo could be inhibited by SAG, suggesting that harmful DDIs could occur when SAG or its medical preparations are co-administered with drugs primarily cleared by these CYP isoforms. sanguinarine 160-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 23500771-7 2013 In vitro-in vivo extrapolation (IV-IVE) from HLM data showed that more than 35.9% of CYP1A2, CYP2C9, CYP2C8 and CYP3A4 activities in vivo could be inhibited by SAG, suggesting that harmful DDIs could occur when SAG or its medical preparations are co-administered with drugs primarily cleared by these CYP isoforms. sanguinarine 211-214 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 93-99 23657428-4 2013 There is overwhelming evidence that an individual"s warfarin maintenance is associated with clinical factors and genetic variations, most notably polymorphisms in cytochrome P450 2C9 and vitamin K epoxide reductase subunit 1. Warfarin 52-60 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-182 23588782-15 2013 CYP2C9 is responsible for metabolising vitamin K antagonists (VKAs), non-steroidal anti-inflammatory drugs (NSAIDs), sulfonylureas, angiotensin II receptor antagonists and phenytoin. Vitamin K 39-48 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23588782-15 2013 CYP2C9 is responsible for metabolising vitamin K antagonists (VKAs), non-steroidal anti-inflammatory drugs (NSAIDs), sulfonylureas, angiotensin II receptor antagonists and phenytoin. Sulfonylurea Compounds 117-130 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23588782-15 2013 CYP2C9 is responsible for metabolising vitamin K antagonists (VKAs), non-steroidal anti-inflammatory drugs (NSAIDs), sulfonylureas, angiotensin II receptor antagonists and phenytoin. Phenytoin 172-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23588782-19 2013 A relationship between CYP2C9 polymorphisms and the pharmacokinetics of sulfonylureas and angiotensin II receptor antagonists has also been observed. Sulfonylurea Compounds 72-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 23-29 23588782-21 2013 Finally, homozygous and heterozygous carriers of CYP2C9 variant alleles require lower doses of phenytoin to reach therapeutic plasma concentrations, and are at increased risk of toxicity. Phenytoin 95-104 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 23602689-1 2013 The aim of this study was to determine the frequencies of SNPs in the vitamin K epoxide reductase complex subunit 1 (VKORC1) and cytochrome P450 2C9 (CYP2C9) genes and their effect on warfarin dose requirement, over anticoagulation and other adverse outcomes in Indian population. Warfarin 184-192 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 150-156 23602689-2 2013 A total of 145 warfarin treated patients for various clinical conditions were screened for VKORC1 and CYP2C9 gene polymorphisms by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique. Warfarin 15-23 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 102-108 23201087-0 2013 Effect of different genetics variants: CYP2C9*2, CYP2C9*3 of cytochrome P-450 CYP2C9 and 1639G>A of the VKORC1 gene; On acenocoumarol requirement in Moroccan patients. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 39-45 23201087-0 2013 Effect of different genetics variants: CYP2C9*2, CYP2C9*3 of cytochrome P-450 CYP2C9 and 1639G>A of the VKORC1 gene; On acenocoumarol requirement in Moroccan patients. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 23201087-0 2013 Effect of different genetics variants: CYP2C9*2, CYP2C9*3 of cytochrome P-450 CYP2C9 and 1639G>A of the VKORC1 gene; On acenocoumarol requirement in Moroccan patients. Acenocoumarol 123-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 49-55 23582453-0 2013 Implication of novel CYP2C9*57 (p.Asn204His) variant in coumarin hypersensitivity. coumarin 56-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 23582453-5 2013 The pharmacoproteomic significance of the novel genetic variant was elucidated by structural demonstration of binding of coumarin molecules within the mutant CYP2C9 204His protein model and in silico bioinformatic evolutionary analyses. coumarin 121-129 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 158-164 23666577-4 2013 O-demethylation is catalyzed by the highly polymorphic CYP2D6 and N-demethylation by several enzymes, CYP2C19, CYP2C9, and CYP3A4. Nitrogen 66-67 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 111-117 23582453-10 2013 Analysis of 3D structure model of the mutant protein revealed that the substituted His204 led to restricted binding of the coumarin drug within the binding site of CYP2C9 enzyme, thereby inhibiting its metabolic clearance and thus explaining the enhanced pharmacologic effect and bleeding in the patient. coumarin 123-131 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 164-170 23666577-5 2013 The observed overall pharmacokinetic effect was most probably the result of decreased N-demethylation of venlafaxine by (1) reduced expression of CYP2C19 due to a genetic deficit and (2) inhibition of CYP2C9 by cotrimoxazole. Nitrogen 86-87 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 23582453-11 2013 CONCLUSIONS: The study elucidates the structurally deleterious role of the novel CYP2C9*57 missense mutation in coumarin toxicity. coumarin 112-120 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 81-87 23666577-5 2013 The observed overall pharmacokinetic effect was most probably the result of decreased N-demethylation of venlafaxine by (1) reduced expression of CYP2C19 due to a genetic deficit and (2) inhibition of CYP2C9 by cotrimoxazole. Venlafaxine Hydrochloride 105-116 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 23641937-4 2013 Here we present a comprehensive study of the oxidation of three widely used pharmaceutical compounds (S-ibuprofen, diclofenac, and S-warfarin) by one of the major drug-metabolizing P450 isoforms, CYP2C9. Sulfur 102-103 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 23666577-5 2013 The observed overall pharmacokinetic effect was most probably the result of decreased N-demethylation of venlafaxine by (1) reduced expression of CYP2C19 due to a genetic deficit and (2) inhibition of CYP2C9 by cotrimoxazole. Trimethoprim, Sulfamethoxazole Drug Combination 211-224 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 201-207 23201087-6 2013 The results showed that the allelic frequencies of the three mutations studied was varies, most of patients having CYP2C9*2 and CYP2C9*3 mutations belong to a group with low dose of acenocoumarol, with P-value of 0.0082 and the single patient with CYP2C9*3 on homozygous form belongs to the same group and carried the A allele for VKORC1 gene. Acenocoumarol 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 115-121 23201087-6 2013 The results showed that the allelic frequencies of the three mutations studied was varies, most of patients having CYP2C9*2 and CYP2C9*3 mutations belong to a group with low dose of acenocoumarol, with P-value of 0.0082 and the single patient with CYP2C9*3 on homozygous form belongs to the same group and carried the A allele for VKORC1 gene. Acenocoumarol 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 23201087-6 2013 The results showed that the allelic frequencies of the three mutations studied was varies, most of patients having CYP2C9*2 and CYP2C9*3 mutations belong to a group with low dose of acenocoumarol, with P-value of 0.0082 and the single patient with CYP2C9*3 on homozygous form belongs to the same group and carried the A allele for VKORC1 gene. Acenocoumarol 182-195 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 128-134 23641937-4 2013 Here we present a comprehensive study of the oxidation of three widely used pharmaceutical compounds (S-ibuprofen, diclofenac, and S-warfarin) by one of the major drug-metabolizing P450 isoforms, CYP2C9. Ibuprofen 104-113 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 23641937-4 2013 Here we present a comprehensive study of the oxidation of three widely used pharmaceutical compounds (S-ibuprofen, diclofenac, and S-warfarin) by one of the major drug-metabolizing P450 isoforms, CYP2C9. Diclofenac 115-125 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 23641937-4 2013 Here we present a comprehensive study of the oxidation of three widely used pharmaceutical compounds (S-ibuprofen, diclofenac, and S-warfarin) by one of the major drug-metabolizing P450 isoforms, CYP2C9. Sulfur 131-132 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 23641937-4 2013 Here we present a comprehensive study of the oxidation of three widely used pharmaceutical compounds (S-ibuprofen, diclofenac, and S-warfarin) by one of the major drug-metabolizing P450 isoforms, CYP2C9. Warfarin 133-141 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 196-202 23641937-9 2013 The carboxylate of diclofenac interacts strongly with the CYP2C9 Arg108 side chain in the transition state for formation of the observed metabolite-but not in that for the competing pathway. carboxylate 4-15 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 23641937-9 2013 The carboxylate of diclofenac interacts strongly with the CYP2C9 Arg108 side chain in the transition state for formation of the observed metabolite-but not in that for the competing pathway. Diclofenac 19-29 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 58-64 23691226-2 2013 Three polymorphisms in two genes, VKORC1 and CYP2C9, are the main coumarin dose determinants and no additional polymorphisms of any relevant pharmacogenetic importance have been identified. coumarin 66-74 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 23208322-6 2013 RESULTS: Warfarin dose was partially correlated with the VKORC1 rs7294, CYP2C9 rs1057910 and ORM1 rs17650 polymorphisms. Warfarin 9-17 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 27122712-5 2013 Clinical and genetic factors such as polymorphisms in CYP2C9 and VKORC1 associated with an individual"s warfarin maintenance have been identified. Warfarin 104-112 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 23519598-8 2013 RESULTS: The model confirmed CYP2C9 and VKORC1 variants as the major predictors of variability in phenprocoumon concentrations and effects, together with body weight, age, comedication with CYP3A modifiers (i.e. inhibitors or inducers) and presence of atrial fibrillation. Phenprocoumon 98-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-35 23559402-9 2013 Polymorphisms in CYP2C9, TPMT, UGT1A1, MDR1, and 5-HTR2A were related to some adverse effects of olanzapine. Olanzapine 97-107 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 17-23 24324947-0 2013 VKORC1 and CYP2C9 Genotype Variations in Relation to Warfarin Dosing in Korean Stroke Patients. Warfarin 53-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17 24324947-2 2013 We sought to establish a warfarin dosing formula for individualized target International Normalization Ratio of Prothrombin Times (INRs) using data from single nucleotide polymorphisms (SNPs) in VKORC1 and CYP2C9 in Korean patients. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 206-212 23651023-0 2013 Impact of CYP2C9 polymorphisms on the vulnerability to pharmacokinetic drug-drug interactions during acenocoumarol treatment. Acenocoumarol 101-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 22767428-5 2013 CYP3A4, CYP2C9 and CYP1A2 were identified to be the main CYP isoforms involved in the metabolism of evodiamine in human liver microsomes. evodiamine 100-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23651023-1 2013 AIM: The objective of this study was to investigate the impact of CYP2C9 polymorphisms and drug-drug interactions on the risk of overanticoagulation in patients treated with acenocoumarol, a vitamin K antagonist. Acenocoumarol 174-187 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 23651023-1 2013 AIM: The objective of this study was to investigate the impact of CYP2C9 polymorphisms and drug-drug interactions on the risk of overanticoagulation in patients treated with acenocoumarol, a vitamin K antagonist. Vitamin K 191-200 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 66-72 23399640-6 2013 In contrast to PTI, indomethacin is primarily O-demethylated by CYP2C9, which prefers acidic substrates. Indomethacin 20-32 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 22943633-1 2013 AIM: The present study evaluated the possibility of drug interactions involving blueberry juice (BBJ) and substrate drugs whose clearance is dependent on cytochromes P4503A (CYP3A) and P4502C9 (CYP2C9). blueberry juice 80-95 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 22943633-1 2013 AIM: The present study evaluated the possibility of drug interactions involving blueberry juice (BBJ) and substrate drugs whose clearance is dependent on cytochromes P4503A (CYP3A) and P4502C9 (CYP2C9). epacadostat 97-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 194-200 22943633-2 2013 METHODS: A 50:50 mixture of lowbush and highbush BBJ was evaluated in vitro as an inhibitor of CYP3A activity (hydroxylation of triazolam and dealkylation of buspirone) and of CYP2C9 activity (flurbiprofen hydroxylation) using human liver microsomes. epacadostat 49-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 176-182 22943633-4 2013 RESULTS: BBJ inhibited CYP3A and CYP2C9 activity in vitro, with 50% inhibitory concentrations (IC50 ) of less than 2%, but without evidence of mechanism-based (irreversible) inhibition. epacadostat 9-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 33-39 22943633-9 2013 In the study of flurbiprofen (CYP2C9 substrate), the positive control inhibitor fluconazole significantly increased flurbiprofen AUC (GMR = 1.71), but BBJ had no significant effect (GMR = 1.03). Flurbiprofen 16-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 22943633-9 2013 In the study of flurbiprofen (CYP2C9 substrate), the positive control inhibitor fluconazole significantly increased flurbiprofen AUC (GMR = 1.71), but BBJ had no significant effect (GMR = 1.03). Fluconazole 80-91 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 22943633-9 2013 In the study of flurbiprofen (CYP2C9 substrate), the positive control inhibitor fluconazole significantly increased flurbiprofen AUC (GMR = 1.71), but BBJ had no significant effect (GMR = 1.03). Flurbiprofen 116-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 30-36 23845193-6 2013 The complete inhibition of the CYP2C9 and CYP3A4 mediated metabolic pathway of glibenclamide through sorafenib might have resulted in a rapid accumulation of glibenclamide. Glyburide 79-92 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 23845193-6 2013 The complete inhibition of the CYP2C9 and CYP3A4 mediated metabolic pathway of glibenclamide through sorafenib might have resulted in a rapid accumulation of glibenclamide. Sorafenib 101-110 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 23845193-6 2013 The complete inhibition of the CYP2C9 and CYP3A4 mediated metabolic pathway of glibenclamide through sorafenib might have resulted in a rapid accumulation of glibenclamide. Glyburide 158-171 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 31-37 22990331-0 2013 CYP2C9 and VKORC1 polymorphisms influence warfarin dose variability in patients on long-term anticoagulation. Warfarin 42-50 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 22990331-1 2013 OBJECTIVES: The main aim of this study was to determine whether CYP2C9 and VKORC1 polymorphisms influence warfarin dose variability during initial dose-finding phase and during maintenance treatment after 360 days. Warfarin 106-114 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 22990331-4 2013 RESULTS: During the first 30 days of anticoagulation, the relative variability of warfarin dose was significantly associated with CYP2C9*2 and CYP2C9*3 polymorphisms (p = 0.02) and with VKORC1 1639G >A genotypes (p = 0.04). Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 130-136 22990331-4 2013 RESULTS: During the first 30 days of anticoagulation, the relative variability of warfarin dose was significantly associated with CYP2C9*2 and CYP2C9*3 polymorphisms (p = 0.02) and with VKORC1 1639G >A genotypes (p = 0.04). Warfarin 82-90 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 143-149 22990331-8 2013 CONCLUSIONS: Genetic information on CYP2C9 and VKORC1 is important both for the initial dose-finding phase and during maintenance treatment with warfarin. Warfarin 145-153 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 36-42 23183958-2 2013 Single gene polymorphisms in cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) have been shown to impact warfarin dosing in adults. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 29-48 23380426-0 2013 Evaluation of the pharmacokinetic interaction between ticagrelor and tolbutamide, a cytochrome P450 2C9 substrate, in healthy volunteers. Ticagrelor 54-64 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 23380426-0 2013 Evaluation of the pharmacokinetic interaction between ticagrelor and tolbutamide, a cytochrome P450 2C9 substrate, in healthy volunteers. Tolbutamide 69-80 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 84-103 23380426-1 2013 OBJECTIVES: To assess the effect of ticagrelor on the pharmacokinetics of tolbutamide (a CYP2C9 substrate), and the effect of tolbutamide on ticagrelor pharmacokinetics. Tolbutamide 74-85 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 23183958-2 2013 Single gene polymorphisms in cytochrome P450 2C9 (CYP2C9) and vitamin K epoxide reductase (VKORC1) have been shown to impact warfarin dosing in adults. Warfarin 125-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 50-56 23376925-1 2013 Cytochrome P450 2C9 (CYP2C9) c.449G>A (*8) is common in African Americans and is associated with decreased warfarin clearance. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-19 23376925-1 2013 Cytochrome P450 2C9 (CYP2C9) c.449G>A (*8) is common in African Americans and is associated with decreased warfarin clearance. Warfarin 110-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 21-27 23376925-7 2013 These data suggest that promoter region polymorphisms inherited with 449G>A decrease CYP2C9 expression and contribute to CYP2C9*8 effects on warfarin clearance and dose requirements. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 88-94 23376925-7 2013 These data suggest that promoter region polymorphisms inherited with 449G>A decrease CYP2C9 expression and contribute to CYP2C9*8 effects on warfarin clearance and dose requirements. Warfarin 144-152 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 124-130 23556443-0 2013 Combining genetic variations in CYP2C9 and VKORC1 with clinical factors for warfarin dosing determination improved clinical effectiveness. Warfarin 76-84 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 27536444-5 2013 METHODS: The ability of 4-AP to inhibit CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4/5 in a direct and time-dependent manner was evaluated using pooled human liver microsomes. 4-Aminopyridine 24-28 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 23298603-0 2013 The role of CYP2C9 polymorphisms in phenytoin-related cerebellar atrophy. Phenytoin 36-45 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 12-18 23298603-2 2013 It is also known that a CYP2C9 mutation (*2 or *3) reduces phenytoin metabolism by 25-50% and can increase the risk of phenytoin-related side effects. Phenytoin 59-68 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 23298603-2 2013 It is also known that a CYP2C9 mutation (*2 or *3) reduces phenytoin metabolism by 25-50% and can increase the risk of phenytoin-related side effects. Phenytoin 119-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 24-30 23298603-8 2013 CONCLUSION: Our study is the first to report evidence linking CYP2C9 polymorphism and a reduction in cerebellar volume in epileptic users of phenytoin. Phenytoin 141-150 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 62-68 23473641-0 2013 Effect of CYP2C9 and VKORC1 genetic polymorphisms on mean daily maintenance dose of acenocoumarol in South Indian patients. Acenocoumarol 84-97 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 23473641-1 2013 AIM: To determine the effect of CYP2C9 and VKORC1 genetic polymorphisms on mean daily maintenance dose of acenocoumarol in South Indian patients with heart valve replacement. Acenocoumarol 106-119 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 32-38 23473641-4 2013 RESULTS: Patients with at least one variant allele of CYP2C9 (*1*2 or *1*3) required 44% and 28.2% lower daily maintenance dose of acenocoumarol (2.0mg and 2.5mg, respectively) than the normal CYP2C9*1*1 genotype group (3.4mg) (p<0.05). Acenocoumarol 131-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 23473641-4 2013 RESULTS: Patients with at least one variant allele of CYP2C9 (*1*2 or *1*3) required 44% and 28.2% lower daily maintenance dose of acenocoumarol (2.0mg and 2.5mg, respectively) than the normal CYP2C9*1*1 genotype group (3.4mg) (p<0.05). Acenocoumarol 131-144 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 193-199 23473641-7 2013 The clinical (age, body mass index) and genetic variables (VKORC1-1639 G>A, CYP2C9*2, CYP2C9*3) contribute together to predict 30.4% of the required maintenance dose of acenocoumarol. Acenocoumarol 172-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 79-85 23473641-7 2013 The clinical (age, body mass index) and genetic variables (VKORC1-1639 G>A, CYP2C9*2, CYP2C9*3) contribute together to predict 30.4% of the required maintenance dose of acenocoumarol. Acenocoumarol 172-185 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 89-95 23473641-8 2013 CONCLUSION: The genetic polymorphisms of CYP2C9 and VKORC1 results in decreased requirement of daily maintenance dose of acenocoumarol. Acenocoumarol 121-134 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 41-47 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Bupropion 172-181 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Diclofenac 183-193 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23153057-3 2013 The cocktail strategy (in situ incubation) was used to assess the induction of CYP1A2, CYP2B6, CYP2C9 and CYP3A4 by using the recommended probe substrate, i.e. phenacetin, bupropion, diclofenac and midazolam, respectively. Midazolam 198-207 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 95-101 23427827-6 2013 Fitting of the data with the Poole-Frenkel model indicates a correlation between the barrier height for electron transfer and the ease of CYP2C9-mediated metabolism of the bound substrates, though the spin state of iron is not well correlated. Iron 215-219 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 138-144 23376975-2 2013 There is now a large body of published data showing that genotype for certain common polymorphisms in the genes encoding the target vitamin K epoxide reductase (G-1639A/C1173T) and the main metabolizing enzyme CYP2C9 (CYP2C9*2 and *3 alleles) are important determinants of the individual coumarin anticoagulant dose requirement. coumarin 288-296 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 210-216 23376975-2 2013 There is now a large body of published data showing that genotype for certain common polymorphisms in the genes encoding the target vitamin K epoxide reductase (G-1639A/C1173T) and the main metabolizing enzyme CYP2C9 (CYP2C9*2 and *3 alleles) are important determinants of the individual coumarin anticoagulant dose requirement. coumarin 288-296 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 218-224 23061746-8 2013 In our study, in addition to age, genetic variants of CYP2C9, VKORC1 and CYP4F2 were found to be significant predictor variables for the maintenance dose for warfarin, explaining 39.3% of dose variability. Warfarin 158-166 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 54-60 23061746-0 2013 Impact of genetic factors (CYP2C9, VKORC1 and CYP4F2) on warfarin dose requirement in the Turkish population. Warfarin 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 27-33 23061746-3 2013 The main objective of this study was to determine whether and to what extent variability in warfarin dose requirements was determined by polymorphisms in CYP2C9, VKORC1, CYP4F2 (rs2108622) and EPHX1 (rs2292566) in the Turkish population. Warfarin 92-100 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 154-160 23159639-0 2013 The impact of the CYP2C9 and VKORC1 polymorphisms on acenocoumarol dose requirements in a Romanian population. Acenocoumarol 53-66 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 18-24 23159639-10 2013 CONCLUSION: The stable therapeutic dose of acenocoumarol is dependent of patient"s age, the presence of the CYP2C9*3 allele and the c.-1639G>A polymorphism of VKORC1. Acenocoumarol 43-56 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 108-114 23826879-7 2013 Amitifadine was a moderate inhibitor of the human isoforms of the major drug metabolizing enzymes CYP2D6, CYP3A4, CYP2C9, and CYP2C19 (IC50 = 9 - 100 muM), but was a potent inhibitor of human CYP2B6 (IC50 = 1.8 muM). 1-(3,4-dichlorophenyl)-3-azabicyclo-(3.1.0)hexane hydrochloride 0-11 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 24329780-3 2013 The present study extends these findings by testing the hypothesis that CYP2C9- and 1A2-selective chemical inhibitors, sulfaphenazole (SFZ) and alpha-naphthoflavone (ANF), block oxidation of JWH-018 and AM2201 in human liver microsomes (HLM). Sulfaphenazole 119-133 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24329780-3 2013 The present study extends these findings by testing the hypothesis that CYP2C9- and 1A2-selective chemical inhibitors, sulfaphenazole (SFZ) and alpha-naphthoflavone (ANF), block oxidation of JWH-018 and AM2201 in human liver microsomes (HLM). Sulfaphenazole 135-138 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24329780-3 2013 The present study extends these findings by testing the hypothesis that CYP2C9- and 1A2-selective chemical inhibitors, sulfaphenazole (SFZ) and alpha-naphthoflavone (ANF), block oxidation of JWH-018 and AM2201 in human liver microsomes (HLM). alpha-naphthoflavone 144-164 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24329780-3 2013 The present study extends these findings by testing the hypothesis that CYP2C9- and 1A2-selective chemical inhibitors, sulfaphenazole (SFZ) and alpha-naphthoflavone (ANF), block oxidation of JWH-018 and AM2201 in human liver microsomes (HLM). alpha-naphthoflavone 166-169 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 24329780-3 2013 The present study extends these findings by testing the hypothesis that CYP2C9- and 1A2-selective chemical inhibitors, sulfaphenazole (SFZ) and alpha-naphthoflavone (ANF), block oxidation of JWH-018 and AM2201 in human liver microsomes (HLM). 1-pentyl-3-(1-naphthoyl)indole 191-198 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 72-78 22855348-5 2013 The S-warfarin concentration was significantly higher in patients with CYP2C9*1/*3 than in those with *1/*1 (p < 0.05). Warfarin 6-14 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 71-77 22855348-8 2013 CONCLUSION: For low-dose warfarin treatment, the VKORC1-1639 G > A and CYP2C9 genotype variations affected the pharmacokinetics and pharmacodynamics of warfarin, while we could not find significant effects of CYP4F2 or CYP2C19 genotype variations on warfarin (metabolite) concentrations or PT-INR. Warfarin 25-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 22855348-8 2013 CONCLUSION: For low-dose warfarin treatment, the VKORC1-1639 G > A and CYP2C9 genotype variations affected the pharmacokinetics and pharmacodynamics of warfarin, while we could not find significant effects of CYP4F2 or CYP2C19 genotype variations on warfarin (metabolite) concentrations or PT-INR. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 22855348-8 2013 CONCLUSION: For low-dose warfarin treatment, the VKORC1-1639 G > A and CYP2C9 genotype variations affected the pharmacokinetics and pharmacodynamics of warfarin, while we could not find significant effects of CYP4F2 or CYP2C19 genotype variations on warfarin (metabolite) concentrations or PT-INR. Warfarin 155-163 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 74-80 23556337-0 2013 CYP2C9*3(1075A > C), ABCB1 and SLCO1B1 genetic polymorphisms and gender are determinants of inter-subject variability in pitavastatin pharmacokinetics. pitavastatin 124-136 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 23556337-6 2013 Compared to CYP2C9*1/*1 carriers, CYP2C9*1/*3 carriers had higher AUC(0-infinity) and Cmax of pitavastatin acid and AUC(0-infinity) of pitavastatin lactone (P<0.05). 7-(2-Cyclopropyl-4-(4-fluorophenyl)quinolin-3-yl)-3,5-dihydroxyhept-6-enoic acid 94-111 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 23556337-6 2013 Compared to CYP2C9*1/*1 carriers, CYP2C9*1/*3 carriers had higher AUC(0-infinity) and Cmax of pitavastatin acid and AUC(0-infinity) of pitavastatin lactone (P<0.05). pitavastatin 135-155 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 34-40 23102508-4 2013 Miltirone showed moderate inhibition on CYP1A2 (IC(50)=1.73 muM) and CYP2C9 (IC(50)=8.61 muM), and weak inhibition on CYP2D6 (IC(50)=30.20 muM) and CYP3A4 (IC(50)=33.88 muM). miltirone 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 23102508-5 2013 Enzyme kinetic studies showed that miltirone competitively inhibited CYP2C9 (K(i)=1.48 muM), and displayed mixed type inhibitions on CYP1A2, CYP2D6 and CYP3A4 with K(i) values of 3.17 muM, 24.25 muM and 35.09 muM, respectively. miltirone 35-44 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 69-75 23260576-8 2013 The monomethyl and dimethyl analogues were shown to have enhanced activity in terms of cytotoxicity, CYP2C9 inhibitory potency, and antiviral activity (up to 6-fold increased potency) compared to the parent compound, silybin B. Ethane 19-27 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 101-107 22963393-7 2013 Cynomolgus CYP2C19 exhibits higher activity for DFOH than cynomolgus CYP2C9 although this reaction is a marker reaction of human CYP2C9. dfoh 48-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 129-135 22963393-9 2013 Cynomolgus CYP2C76 metabolizes non-CYP2C substrates, 7-ethoxyresorufin (human CYP1A substrate) and bufuralol (human CYP2D6 substrate). ethoxyresorufin 53-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-16 22963393-9 2013 Cynomolgus CYP2C76 metabolizes non-CYP2C substrates, 7-ethoxyresorufin (human CYP1A substrate) and bufuralol (human CYP2D6 substrate). bufuralol 99-108 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-16 22837389-6 2013 Estradiol also increased enzyme activities of CYP2C9 and CYP2E1 without affecting the mRNA expression levels by unknown mechanisms. Estradiol 0-9 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 46-52 23429588-4 2013 Treprostinil is primarily metabolized by cytochrome (CYP) P450 2C8 with minor contribution from CYP2C9. treprostinil 0-12 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 96-102 23429588-5 2013 Sildenafil is metabolized by CYP3A4 with minor contribution from CYP2C9. Sildenafil Citrate 0-10 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 65-71 22842957-0 2013 Effect of CYP2C9 and SLCO1B1 polymorphisms on the pharmacokinetics and pharmacodynamics of nateglinide in healthy Chinese male volunteers. Nateglinide 91-102 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 10-16 22842957-2 2013 Our objective was to assess the association between CYP2C9 and SLCO1B1 polymorphisms and the metabolism of nateglinide in healthy Chinese male volunteers. Nateglinide 107-118 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 52-58 22842957-6 2013 CYP2C9*3 and 521 T>C polymorphisms were the significant predictors of the AUC(0- ) and CL/F of nateglinide (adjusted multiple R(2) = 34 % and 43 %, respectively) according to multiple linear regression analyses, but they have no significant association with changes in the blood glucose-lowering effect of nateglinide. Nateglinide 98-109 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 0-6 22842957-7 2013 CONCLUSIONS: Both SLCO1B1 521 T>C and the CYP2C9*3 polymorphisms can significantly affect the pharmacokinetics of nateglinide, but they could only partially explain the interindividual variability of plasma concentration of nateglinide. Nateglinide 117-128 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 45-51 22855348-0 2013 Responsiveness to low-dose warfarin associated with genetic variants of VKORC1, CYP2C9, CYP2C19, and CYP4F2 in an Indonesian population. Warfarin 27-35 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 80-86 22855348-1 2013 PURPOSE: The aim of this study was to assess the pharmacokinetics and pharmacodynamics of warfarin associated with genetic polymorphisms in VKORC1, CYP2C9, CYP2C19, and CYP4F2 in Indonesian patients treated with low-dose warfarin. Warfarin 90-98 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 148-154 23159358-0 2013 Role of CYP2C9 polymorphism in phenytoin-related metabolic abnormalities and subclinical atherosclerosis in young adult epileptic patients. Phenytoin 31-40 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 8-14 23159358-4 2013 The mean serum total triglyceride and LDL levels were significantly higher in the wild type gene subjects than in the CYP 2C9 polymorphism gene subjects. Triglycerides 21-33 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 118-125 23276529-0 2013 Repeated bleeding complications during therapy with vitamin K antagonists in a patient with the VKORC1*2A and the CYP2C9*3/*3 alleles: genetic testing to support switching to new oral anticoagulants. Vitamin K 52-61 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 114-120 22725836-2 2013 Previous in vitro studies indicate that curcuminoids and piperine (a black pepper derivative that enhances curcuminoid bioavailability) could inhibit human CYP3A, CYP2C9, UGT and SULT dependent drug metabolism. curcuminoids 40-52 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 22725836-2 2013 Previous in vitro studies indicate that curcuminoids and piperine (a black pepper derivative that enhances curcuminoid bioavailability) could inhibit human CYP3A, CYP2C9, UGT and SULT dependent drug metabolism. piperine 57-65 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 22725836-2 2013 Previous in vitro studies indicate that curcuminoids and piperine (a black pepper derivative that enhances curcuminoid bioavailability) could inhibit human CYP3A, CYP2C9, UGT and SULT dependent drug metabolism. curcuminoid 40-51 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 163-169 23206182-2 2013 AIM: To determine the clinical effectiveness of CYP2D6, CYP3A4, CYP2C9/19, and UGT polymorphism on the steady-state plasma concentrations and therapeutic outcome of rivastigmine monotherapy and combination therapy in patients with Alzheimer"s disease. Rivastigmine 165-177 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 64-70 23061746-9 2013 VKORC1 and CYP2C9 genotypes remain predictor variables of the warfarin dose, and we first found that CYP4F2 (rs2108622) contributes to dose variability in the Turkish population as well. Warfarin 62-70 cytochrome P450 family 2 subfamily C member 9 Homo sapiens 11-17