PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 31480671-5 2019 Among the top 10 most probable inhibitors of TRPA1 with good blood brain barrier (BBB) permeability, desvenlafaxine, paliperidone, and febuxostat emerged as the most promising repurposable agents for treating MS. Molecular docking studies indicated that desvenlafaxine, paliperidone, and febuxostat are likely to induce allosteric TRPA1 channel inhibition. Desvenlafaxine Succinate 101-115 transient receptor potential cation channel subfamily A member 1 Homo sapiens 45-50 31480671-5 2019 Among the top 10 most probable inhibitors of TRPA1 with good blood brain barrier (BBB) permeability, desvenlafaxine, paliperidone, and febuxostat emerged as the most promising repurposable agents for treating MS. Molecular docking studies indicated that desvenlafaxine, paliperidone, and febuxostat are likely to induce allosteric TRPA1 channel inhibition. Desvenlafaxine Succinate 254-268 transient receptor potential cation channel subfamily A member 1 Homo sapiens 45-50 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 52-74 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 52-74 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 234-240 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 76-79 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 76-79 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 234-240 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 81-95 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 30838456-7 2019 Venlafaxine is metabolized to the active metabolite O-desmethylvenlafaxine (ODV; desvenlafaxine) by CYP2D6, and it therefore is subject to significant inter-individual variation in blood levels and response dependent on variations in CYP2D6 metabolism. Desvenlafaxine Succinate 81-95 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 234-240 29732526-8 2018 Alpha-lipoic acid and DVS, alone or combined, reversed CORT effect on TST. Desvenlafaxine Succinate 22-25 thiosulfate sulfurtransferase Homo sapiens 70-73 28079059-8 2017 Changes in miR-1202 levels following desvenlafaxine treatment were negatively correlated with changes in activity in right precuneus within the default-mode network, and in connectivity between the posterior cingulate and the temporal and prefrontal cortices, and the precuneus. Desvenlafaxine Succinate 37-51 microRNA 1202 Homo sapiens 11-19 29897361-3 2018 Venlafaxine and its metabolite O-desmethylvenlafaxine were the primary compounds (reaching up to 132.04 and 173.68 ng L-1 as well as 3.03 and 4.53 ng L-1 in wastewater and receiving water, respectively). Desvenlafaxine Succinate 31-53 L1 cell adhesion molecule Homo sapiens 118-121 29897361-3 2018 Venlafaxine and its metabolite O-desmethylvenlafaxine were the primary compounds (reaching up to 132.04 and 173.68 ng L-1 as well as 3.03 and 4.53 ng L-1 in wastewater and receiving water, respectively). Desvenlafaxine Succinate 31-53 L1 cell adhesion molecule Homo sapiens 150-153 28866861-10 2018 The findings might be the result of different degrees of CYP2C19 involvement, therefore the inhibition of CYP2C19 by both PPIs may lead to an increased metabolism via CYP2D6 to O-desmethylvenlafaxine. Desvenlafaxine Succinate 177-199 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 57-64 28866861-10 2018 The findings might be the result of different degrees of CYP2C19 involvement, therefore the inhibition of CYP2C19 by both PPIs may lead to an increased metabolism via CYP2D6 to O-desmethylvenlafaxine. Desvenlafaxine Succinate 177-199 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 106-113 28866861-10 2018 The findings might be the result of different degrees of CYP2C19 involvement, therefore the inhibition of CYP2C19 by both PPIs may lead to an increased metabolism via CYP2D6 to O-desmethylvenlafaxine. Desvenlafaxine Succinate 177-199 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 167-173 26331791-3 2016 Venlafaxine (VLX) is an antidepressant metabolized mainly by CYP2D6 to O-desmethylvenlafaxine (ODV), CYP3A to N-desmethylvenlafaxine (NDV), and CYP2D6 and CYP3A to N,O-didesmethylvenlafaxine (DDV). Desvenlafaxine Succinate 71-93 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 61-67 27696548-0 2017 Spectroscopic exploration and thermodynamic characterization of desvenlafaxine interacting with fluorescent bovine serum albumin. Desvenlafaxine Succinate 64-78 albumin Homo sapiens 115-128 27696548-1 2017 The mechanism of the interaction between bovine serum albumin (BSA) and desvenlafaxine was studied using fluorescence, ultraviolet absorption, 3-dimensional fluorescence spectroscopy, circular dichroism, synchronous fluorescence spectroscopy, cyclic voltametry, differential scanning calorimetry, and attenuated total reflection-Fourier transform infrared spectroscopic techniques under physiological condition at pH 7.4. Desvenlafaxine Succinate 72-86 albumin Homo sapiens 48-61 27328779-2 2016 In humans, venlafaxine is biotransformed for the most part by CYP2D6 and CYP2C19 isoenzymes to its major metabolite O-desmethylvenlafaxine (ODV), and in parallel to N-desmethylvenlafaxine (NDV) and N,O-didesmethylvenlafaxine (NODV) by several CYP isoenzymes, mainly including CYP3A4 and CYP2C19. Desvenlafaxine Succinate 116-138 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 73-80 28465217-4 2017 This study evaluates the involvement of nine common antidepressants in the NLRP3-inflammasome complex (fluoxetine, paroxetine, mianserin, mirtazapine, venlafaxine, desvenlafaxine, amitriptyline, imipramine and agomelatine), both in in vitro THP-1 cells stimulated by ATP, and in a stress-induced depressive animal or MDD patients. Desvenlafaxine Succinate 164-178 NLR family pyrin domain containing 3 Homo sapiens 75-80 28229443-5 2017 CONCLUSIONS: Although evidence of desvenlafaxine in neuropathic pain is scarce, it presents some interesting pharmacokinetic properties, as it is not substrate or have activity on P-glycoprotein, and have a metabolism which practically does not depend on cytochrome P450 system, which limits the risk of pharmacokinetic interactions and potential problems associated tolerability when administered with drugs that are CYP2D6 moderate or potent inhibitors or other substrates of this isoenzyme. Desvenlafaxine Succinate 34-48 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 418-424 29123929-3 2017 VEN is metabolized mainly by CYP2D6 and to a minor extent by CYP2C19 to the major active metabolite O-desmethylvenlafaxine (ODV). Desvenlafaxine Succinate 100-122 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 61-68 27023264-1 2016 No studies to date have evaluated SLC6A2 and SLC6A4 genetic polymorphisms influencing antidepressant response to desvenlafaxine. Desvenlafaxine Succinate 113-127 solute carrier family 6 member 4 Homo sapiens 45-51 27023264-2 2016 We conducted an 8-week, open-label, prospective pilot study in 35 patients with major depressive disorder to assess the effects of genetic variations in SLC6A2 and SLC6A4 on both efficacy and side effect profile of desvenlafaxine. Desvenlafaxine Succinate 215-229 solute carrier family 6 member 2 Homo sapiens 153-159 27023264-2 2016 We conducted an 8-week, open-label, prospective pilot study in 35 patients with major depressive disorder to assess the effects of genetic variations in SLC6A2 and SLC6A4 on both efficacy and side effect profile of desvenlafaxine. Desvenlafaxine Succinate 215-229 solute carrier family 6 member 4 Homo sapiens 164-170 27023264-5 2016 While these results need to be interpreted cautiously, they provide preliminary support for the SLC6A4 HTTLPR polymorphism as potential modifier of desvenlafaxine efficacy. Desvenlafaxine Succinate 148-162 solute carrier family 6 member 4 Homo sapiens 96-102 26331791-3 2016 Venlafaxine (VLX) is an antidepressant metabolized mainly by CYP2D6 to O-desmethylvenlafaxine (ODV), CYP3A to N-desmethylvenlafaxine (NDV), and CYP2D6 and CYP3A to N,O-didesmethylvenlafaxine (DDV). Desvenlafaxine Succinate 95-98 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 61-67 23541126-5 2013 CYP2D6 poor metabolizer phenotype was defined as O-desmethylvenlafaxine to venlafaxine ratio < 1 based on published data. Desvenlafaxine Succinate 49-71 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 26514447-0 2015 Comparison of efficacy, safety and brain derived neurotrophic factor (BDNF) levels in patients of major depressive disorder, treated with fluoxetine and desvenlafaxine. Desvenlafaxine Succinate 153-167 brain derived neurotrophic factor Homo sapiens 70-74 26514447-1 2015 This randomized, open label, prospective, observational study compared clinical efficacy, safety alongwith plasma BDNF levels in outpatients of depression treated with fluoxetine and desvenlafaxine. Desvenlafaxine Succinate 183-197 brain derived neurotrophic factor Homo sapiens 114-118 26514447-7 2015 The BDNF levels in the desvenlafaxine group were 760.5+-28.53pg/ml at the start of treatment which significantly (p<0.05) increased to 845.8+-32.82pg/ml at 12 weeks. Desvenlafaxine Succinate 23-37 brain derived neurotrophic factor Homo sapiens 4-8 26091569-6 2015 Desvenlafaxine in vivo dissolution from the IRF and ERF was predicted from in vitro solubility studies and biorelevant dissolution studies (using the USP3 dissolution apparatus), respectively. Desvenlafaxine Succinate 0-14 ubiquitin specific peptidase 3 Homo sapiens 150-154 23897419-1 2013 Venlafaxine, and to a lesser extent desvenlafaxine, has previously been shown to induce the expression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in whole cells and alter the cellular permeability of a known drug efflux probe (rhodamine 123). Desvenlafaxine Succinate 36-50 phosphoglycolate phosphatase Mus musculus 135-149 23897419-1 2013 Venlafaxine, and to a lesser extent desvenlafaxine, has previously been shown to induce the expression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in whole cells and alter the cellular permeability of a known drug efflux probe (rhodamine 123). Desvenlafaxine Succinate 36-50 phosphoglycolate phosphatase Mus musculus 151-155 23897419-1 2013 Venlafaxine, and to a lesser extent desvenlafaxine, has previously been shown to induce the expression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in whole cells and alter the cellular permeability of a known drug efflux probe (rhodamine 123). Desvenlafaxine Succinate 36-50 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 161-193 23897419-1 2013 Venlafaxine, and to a lesser extent desvenlafaxine, has previously been shown to induce the expression of the drug efflux transporters P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in whole cells and alter the cellular permeability of a known drug efflux probe (rhodamine 123). Desvenlafaxine Succinate 36-50 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 195-199 26350703-0 2015 Reversal of corticosterone-induced BDNF alterations by the natural antioxidant alpha-lipoic acid alone and combined with desvenlafaxine: Emphasis on the neurotrophic hypothesis of depression. Desvenlafaxine Succinate 121-135 brain derived neurotrophic factor Mus musculus 35-39 24096053-13 2014 Although median increase in BDNF was not significantly different between desvenlafaxine (13.7%) and placebo (5.7%) groups, the increase was significantly greater (33.4% vs 4.3%; P=0.003) in patients with more severe depression at baseline (HAM-D17>22) vs those with less severe depression (HAM-D17<=22). Desvenlafaxine Succinate 73-87 brain derived neurotrophic factor Homo sapiens 28-32 23897419-3 2013 P-gp and BCRP expression was significantly upregulated in the intestine, following a treatment with venlafaxine (2.6- and 6.7-fold, respectively) or desvenlafaxine (2.3- and 4.8-fold, respectively). Desvenlafaxine Succinate 149-163 phosphoglycolate phosphatase Mus musculus 0-4 23897419-3 2013 P-gp and BCRP expression was significantly upregulated in the intestine, following a treatment with venlafaxine (2.6- and 6.7-fold, respectively) or desvenlafaxine (2.3- and 4.8-fold, respectively). Desvenlafaxine Succinate 149-163 ATP binding cassette subfamily G member 2 (Junior blood group) Mus musculus 9-13 21840145-1 2012 Venlafaxine (VEN) is an antidepressant drug mainly metabolized by the cytochrome P450 (CYP) enzyme CYP2D6 to the active metabolite O-desmethylvenlafaxine (ODV). Desvenlafaxine Succinate 131-153 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 99-105 23221858-1 2013 The results of two single-center, two-period, open-label trials that evaluated the effects of multiple doses of desvenlafaxine on the pharmacokinetics of desipramine, a cytochrome P450 (CYP) 2D6 enzyme substrate, are presented. Desvenlafaxine Succinate 112-126 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 169-194 21840145-1 2012 Venlafaxine (VEN) is an antidepressant drug mainly metabolized by the cytochrome P450 (CYP) enzyme CYP2D6 to the active metabolite O-desmethylvenlafaxine (ODV). Desvenlafaxine Succinate 155-158 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 99-105 21119521-3 2011 In this study, we report on five patients who had a historical pattern suggestive of tolerance to other antidepressant drugs, whose genotyping test indicated that they were CYP2C19 rapid metabolizers, and who experienced "poop out" again when commenced on desvenlafaxine. Desvenlafaxine Succinate 256-270 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 173-180 21925241-3 2011 TP1 was designed as a prodrug of desvenlafaxine. Desvenlafaxine Succinate 33-47 transition protein 1 Homo sapiens 0-3 21925241-6 2011 TP1 (0.06 mmol/kg, orally) rapidly penetrated rat brain and hypothalamus, translated into desvenlafaxine within 1 h, and demonstrated higher bioavailability and better pharmacokinetic properties than desvenlafaxine succinate (DVS). Desvenlafaxine Succinate 90-104 transition protein 1 Rattus norvegicus 0-3 21925241-6 2011 TP1 (0.06 mmol/kg, orally) rapidly penetrated rat brain and hypothalamus, translated into desvenlafaxine within 1 h, and demonstrated higher bioavailability and better pharmacokinetic properties than desvenlafaxine succinate (DVS). Desvenlafaxine Succinate 226-229 transition protein 1 Rattus norvegicus 0-3 21446053-3 2011 The present study expanded upon these findings by examining the effect of venlafaxine and desvenlafaxine on the expression of both P-gp and the breast cancer resistance protein (BCRP) in human brain endothelial cells (HBMEC), an in vitro model of the blood-brain barrier (BBB). Desvenlafaxine Succinate 90-104 ATP binding cassette subfamily B member 1 Homo sapiens 131-135 21446053-3 2011 The present study expanded upon these findings by examining the effect of venlafaxine and desvenlafaxine on the expression of both P-gp and the breast cancer resistance protein (BCRP) in human brain endothelial cells (HBMEC), an in vitro model of the blood-brain barrier (BBB). Desvenlafaxine Succinate 90-104 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 144-176 21446053-3 2011 The present study expanded upon these findings by examining the effect of venlafaxine and desvenlafaxine on the expression of both P-gp and the breast cancer resistance protein (BCRP) in human brain endothelial cells (HBMEC), an in vitro model of the blood-brain barrier (BBB). Desvenlafaxine Succinate 90-104 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 178-182 21099743-6 2011 The sum of venlafaxine plus ODV concentration showed the same pattern as venlafaxine concentrations with CYP2D6 and CYP2C19 genotypes with higher activity scores being associated with a lower venlafaxine plus ODV concentration (2D6 P = 0.01; 2C19 P < 0.001). Desvenlafaxine Succinate 209-212 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 116-123 21099743-7 2011 Because allelic variants in both CYP2D6 and CYP2C19 influence the total concentration of the active compounds venlafaxine and ODV, both CYP2D6 and CYP2C19 genotypes should be considered when using pharmacogenomic information for venlafaxine dose alterations. Desvenlafaxine Succinate 126-129 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 33-39 21099743-1 2011 This study examines the relationship between blood concentrations of venlafaxine and its active metabolite, O-desmethyl venlafaxine (ODV), and genetic variants of the cytochrome P450 enzymes CYP2D6 and CYP2C19 in human subjects. Desvenlafaxine Succinate 108-131 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 191-197 21099743-1 2011 This study examines the relationship between blood concentrations of venlafaxine and its active metabolite, O-desmethyl venlafaxine (ODV), and genetic variants of the cytochrome P450 enzymes CYP2D6 and CYP2C19 in human subjects. Desvenlafaxine Succinate 133-136 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 191-197 21099743-7 2011 Because allelic variants in both CYP2D6 and CYP2C19 influence the total concentration of the active compounds venlafaxine and ODV, both CYP2D6 and CYP2C19 genotypes should be considered when using pharmacogenomic information for venlafaxine dose alterations. Desvenlafaxine Succinate 126-129 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 44-51 21099743-7 2011 Because allelic variants in both CYP2D6 and CYP2C19 influence the total concentration of the active compounds venlafaxine and ODV, both CYP2D6 and CYP2C19 genotypes should be considered when using pharmacogenomic information for venlafaxine dose alterations. Desvenlafaxine Succinate 126-129 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 147-154 21067460-6 2010 Desvenlafaxine has clinically insignificant effects on the activity of CYP and P-gp. Desvenlafaxine Succinate 0-14 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 20441720-1 2010 INTRODUCTION: Venlafaxine, a serotonin-norepinephrine reuptake inhibitor antidepressant, is metabolized primarily by the cytochrome P450 2D6 enzyme into O-desmethylvenlafaxine (ODV). Desvenlafaxine Succinate 153-175 cytochrome P450 2D6 Homo sapiens 121-140 20441720-1 2010 INTRODUCTION: Venlafaxine, a serotonin-norepinephrine reuptake inhibitor antidepressant, is metabolized primarily by the cytochrome P450 2D6 enzyme into O-desmethylvenlafaxine (ODV). Desvenlafaxine Succinate 177-180 cytochrome P450 2D6 Homo sapiens 121-140 19919295-21 2010 The metabolic profile of desvenlafaxine suggests a low risk of drug-drug interactions owing to minimal inhibitory effects on CYP2D6, lack of interaction with p-glycoprotein, and low protein binding. Desvenlafaxine Succinate 25-39 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 125-131 20466523-6 2010 One to six hours post-dose, the brain concentrations of venlafaxine, O-desmethylvenlafaxine and N-desmethylvenlafaxine were 2-3, 2-6 and 3-12 times higher in abcb1ab (-/-) mice compared to abcb1ab (+/+) mice, respectively. Desvenlafaxine Succinate 69-91 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 158-163 19919295-21 2010 The metabolic profile of desvenlafaxine suggests a low risk of drug-drug interactions owing to minimal inhibitory effects on CYP2D6, lack of interaction with p-glycoprotein, and low protein binding. Desvenlafaxine Succinate 25-39 ATP binding cassette subfamily B member 1 Homo sapiens 158-172 19593180-4 2009 The current analysis evaluates the use of the O-desmethylvenlafaxine-venlafaxine ratio (ODV/VEN) after administration of VEN, a CYP2D6 substrate, for determining CYP2D6 metabolic phenotype in healthy adults receiving VEN. Desvenlafaxine Succinate 46-68 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 128-134 21391412-2 2010 However, the influence of the serotonin and noradrenalin reuptake inhibitor (SNRI) venlafaxine and its metabolite O-desmethylvenlafaxine on the stimulated blood cell secretion of IFN-gamma has not been studied so far. Desvenlafaxine Succinate 114-136 interferon gamma Homo sapiens 179-188 20098230-2 2010 The desvenlafaxine/venlafaxine ratio, either after a single dose or at steady state, can be used to determine whether a patient is functionally (i.e., phenotypically) a CYP 2D6 extensive or poor metabolizer (EM or PM). Desvenlafaxine Succinate 4-18 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 169-176 19593180-4 2009 The current analysis evaluates the use of the O-desmethylvenlafaxine-venlafaxine ratio (ODV/VEN) after administration of VEN, a CYP2D6 substrate, for determining CYP2D6 metabolic phenotype in healthy adults receiving VEN. Desvenlafaxine Succinate 46-68 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 162-168 19593180-4 2009 The current analysis evaluates the use of the O-desmethylvenlafaxine-venlafaxine ratio (ODV/VEN) after administration of VEN, a CYP2D6 substrate, for determining CYP2D6 metabolic phenotype in healthy adults receiving VEN. Desvenlafaxine Succinate 88-91 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 128-134 19593180-4 2009 The current analysis evaluates the use of the O-desmethylvenlafaxine-venlafaxine ratio (ODV/VEN) after administration of VEN, a CYP2D6 substrate, for determining CYP2D6 metabolic phenotype in healthy adults receiving VEN. Desvenlafaxine Succinate 88-91 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 162-168 19557107-7 2009 Desvenlafaxine is excreted in urine, it is minimally metabolized via the CYP450 pathway, and is a weak inhibitor of CYP2D6. Desvenlafaxine Succinate 0-14 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 116-122 19127490-3 2009 Unlike various other antidepressants including venlafaxine, desvenlafaxine is not metabolized by cytochrome p450 (CYP) enzyme pathways and is associated with minimal inhibition of CYP enzymes. Desvenlafaxine Succinate 60-74 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 180-183 19629022-0 2009 Desvenlafaxine and venlafaxine exert minimal in vitro inhibition of human cytochrome P450 and P-glycoprotein activities. Desvenlafaxine Succinate 0-14 ATP binding cassette subfamily B member 1 Homo sapiens 94-108 18809731-0 2008 An assessment of drug-drug interactions: the effect of desvenlafaxine and duloxetine on the pharmacokinetics of the CYP2D6 probe desipramine in healthy subjects. Desvenlafaxine Succinate 55-69 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 116-122 19001559-0 2009 The effects of desvenlafaxine and paroxetine on the pharmacokinetics of the cytochrome P450 2D6 substrate desipramine in healthy adults. Desvenlafaxine Succinate 15-29 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 76-95 19001559-1 2009 The potential for cytochrome P450 (CYP) 2D6 substrates to interact with desvenlafaxine (administered as desvenlafaxine succinate) and paroxetine was evaluated. Desvenlafaxine Succinate 72-86 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 18-43 19001559-1 2009 The potential for cytochrome P450 (CYP) 2D6 substrates to interact with desvenlafaxine (administered as desvenlafaxine succinate) and paroxetine was evaluated. Desvenlafaxine Succinate 104-128 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 18-43 19629022-6 2009 CONCLUSIONS: Considering in vitro and available clinical data, desvenlafaxine and venlafaxine appear to have low potential for pharmacokinetic drug-drug interactions via inhibiting the metabolic clearance of concomitant drugs that are substrates of various CYP enzymes, in particular CYP2D6. Desvenlafaxine Succinate 63-77 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 257-260 19629022-6 2009 CONCLUSIONS: Considering in vitro and available clinical data, desvenlafaxine and venlafaxine appear to have low potential for pharmacokinetic drug-drug interactions via inhibiting the metabolic clearance of concomitant drugs that are substrates of various CYP enzymes, in particular CYP2D6. Desvenlafaxine Succinate 63-77 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 284-290 18806899-4 2008 Desvenlafaxine has linear pharmacokinetics, low protein binding, a half-life of approximately 10 hours and is metabolized primarily via glucuronidation, and to a minor extent through CYP3A4. Desvenlafaxine Succinate 0-14 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 183-189 19057238-0 2008 Effect of desvenlafaxine on the cytochrome P450 2D6 enzyme system. Desvenlafaxine Succinate 10-24 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-51 19057238-3 2008 METHODS: Five studies assessing the interaction between desvenlafaxine and CYP2D6 are reviewed. Desvenlafaxine Succinate 56-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 75-81 19057238-10 2008 CONCLUSIONS: Based on the findings presented here, desvenlafaxine is expected to have a low risk for variability in efficacy and safety/tolerability resulting from CYP2D6 polymorphisms or drug-drug interactions when coadministered with CYP2D6 substrates or inhibitors. Desvenlafaxine Succinate 51-65 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 164-170 19057238-10 2008 CONCLUSIONS: Based on the findings presented here, desvenlafaxine is expected to have a low risk for variability in efficacy and safety/tolerability resulting from CYP2D6 polymorphisms or drug-drug interactions when coadministered with CYP2D6 substrates or inhibitors. Desvenlafaxine Succinate 51-65 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 236-242 18698015-11 2008 The decreased potential of CYP2D6 activity with desvenlafaxine compared with the parent drug may be a potential advantage in patients on other medications metabolized via this enzymatic pathway. Desvenlafaxine Succinate 48-62 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 27-33 17673606-0 2007 Desvenlafaxine succinate identifies novel antagonist binding determinants in the human norepinephrine transporter. Desvenlafaxine Succinate 0-24 solute carrier family 6 member 2 Homo sapiens 87-113 17673606-1 2007 Desvenlafaxine succinate (DVS) is a recently introduced antagonist of the human norepinephrine and serotonin transporters (hNET and hSERT, respectively), currently in clinical development for use in the treatment of major depressive disorder and vasomotor symptoms associated with menopause. Desvenlafaxine Succinate 0-24 solute carrier family 6 member 2 Homo sapiens 123-127 17673606-1 2007 Desvenlafaxine succinate (DVS) is a recently introduced antagonist of the human norepinephrine and serotonin transporters (hNET and hSERT, respectively), currently in clinical development for use in the treatment of major depressive disorder and vasomotor symptoms associated with menopause. Desvenlafaxine Succinate 0-24 solute carrier family 6 member 4 Homo sapiens 132-137 17673606-1 2007 Desvenlafaxine succinate (DVS) is a recently introduced antagonist of the human norepinephrine and serotonin transporters (hNET and hSERT, respectively), currently in clinical development for use in the treatment of major depressive disorder and vasomotor symptoms associated with menopause. Desvenlafaxine Succinate 26-29 solute carrier family 6 member 2 Homo sapiens 123-127 17673606-1 2007 Desvenlafaxine succinate (DVS) is a recently introduced antagonist of the human norepinephrine and serotonin transporters (hNET and hSERT, respectively), currently in clinical development for use in the treatment of major depressive disorder and vasomotor symptoms associated with menopause. Desvenlafaxine Succinate 26-29 solute carrier family 6 member 4 Homo sapiens 132-137 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 138-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 138-160 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 138-160 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 83-89 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 54-69 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 71-74 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 83-89 16958828-2 2006 It is metabolized primarily by the highly polymorphic cytochrome P450 (CYP) enzyme CYP2D6 to yield a pharmacologically active metabolite, O-desmethylvenlafaxine (ODV), and to a lesser extent by CYP3A4, to yield N-desmethylvenlafaxine (NDV). Desvenlafaxine Succinate 162-165 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 194-200 24930414-6 2003 Although there is insufficient evidence, a substantial inhibition of CYP 3A3/4 by venlafaxine could result in a meaningful increase in plasma levels of venlafaxine, O-desmethylvenlafaxine, alprazolam and diazepam, particularly in patients who are CYP 2D6 deficient. Desvenlafaxine Succinate 165-187 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 69-76 16642541-3 2006 We investigated the relationship between the CYP2D6 polymorphism and steady-state plasma concentration of venlafaxine (VEN) and its primary metabolite o-desmethylvenlafaxine (ODV) in elderly participants receiving venlafaxine-XR for major depression in order to explore the contribution of pharmacogenetics to medication tolerability. Desvenlafaxine Succinate 151-173 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 45-51 12649369-5 2003 All antidepressants tested except O-desmethylvenlafaxine showed Pgp inhibitory activity with sertraline, desmethylsertraline, and paroxetine being the most potent, comparable with the well known Pgp inhibitor quinidine. Desvenlafaxine Succinate 34-56 ATP binding cassette subfamily B member 1 Homo sapiens 64-67 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 35363936-8 2022 Moreover, IHC analysis of maternal and fetal rat brains showed that groups treated with desvenlafaxine demonstrated a significant increase of NGF protein immunoreactivity compared with that in the controls. Desvenlafaxine Succinate 88-102 nerve growth factor Rattus norvegicus 142-145 34855082-3 2021 JNK inhibitor added to the liver homogenate containing antidepressant enhanced and accelerated the formation of the only pharmacologically active venlafaxine metabolite O-desmethylvenlafaxine in the cell suspension. Desvenlafaxine Succinate 169-191 mitogen-activated protein kinase 8 Homo sapiens 0-3 34197885-7 2021 Children of DVs ages 2-3 had significantly worse dentition (13.9% vs. 4.8%, P = 0.03) and more EBP {least square means (lsmeans) 54.31 vs. 47.59, P = 0.02}. Desvenlafaxine Succinate 12-15 EBP cholestenol delta-isomerase Homo sapiens 95-98 34197885-9 2021 Among children 4-11, male children of DVs had significantly more TBP (lsmeans 70.68 vs. 57.34, P = 0.003), IBP (lsmeans 63.59 vs. 56.16, P = 0.002) and EBP (lsmeans 61.60 vs. 52.93, P = 0.03), but female children did not. Desvenlafaxine Succinate 38-41 DEF6 guanine nucleotide exchange factor Homo sapiens 107-110 34197885-9 2021 Among children 4-11, male children of DVs had significantly more TBP (lsmeans 70.68 vs. 57.34, P = 0.003), IBP (lsmeans 63.59 vs. 56.16, P = 0.002) and EBP (lsmeans 61.60 vs. 52.93, P = 0.03), but female children did not. Desvenlafaxine Succinate 38-41 EBP cholestenol delta-isomerase Homo sapiens 152-155 34197885-10 2021 For children ages 12-18, male children of DVs had more EBP (lsmeans 63.73 vs. 43.51, P = 0.008), while female children of DVs had fewer EBP (lsmeans 45.50 vs. 50.48, P = 0.02). Desvenlafaxine Succinate 42-45 EBP cholestenol delta-isomerase Homo sapiens 55-58 34197885-10 2021 For children ages 12-18, male children of DVs had more EBP (lsmeans 63.73 vs. 43.51, P = 0.008), while female children of DVs had fewer EBP (lsmeans 45.50 vs. 50.48, P = 0.02). Desvenlafaxine Succinate 42-45 EBP cholestenol delta-isomerase Homo sapiens 136-139 34197885-10 2021 For children ages 12-18, male children of DVs had more EBP (lsmeans 63.73 vs. 43.51, P = 0.008), while female children of DVs had fewer EBP (lsmeans 45.50 vs. 50.48, P = 0.02). Desvenlafaxine Succinate 122-125 EBP cholestenol delta-isomerase Homo sapiens 136-139 33204067-9 2020 Compared with that in the control group (without vonoprazan), the pharmacokinetic parameters of venlafaxine and its metabolite, O-desmethylvenlafaxine, were significantly increased in both 5 and 20 mg/kg vonoprazan groups, with an increase in MRO-desmethylvenlafaxine. Desvenlafaxine Succinate 128-150 maestro Rattus norvegicus 243-246 33325920-5 2021 Exploring a multidimensional parameter space spanning cell-adhesive ligand, seeding density, fiber alignment, and stiffness, we found that fibronectin-functionalized DVS matrices composed of highly aligned fibers with low stiffness optimally promoted the organization of functional iPSC-CM tissues. Desvenlafaxine Succinate 166-169 fibronectin 1 Homo sapiens 139-150 31658948-7 2019 In addition, O-desmethyl venlafaxine formation was reduced as well in CYP2D1-null rats compared with that in WT rats. Desvenlafaxine Succinate 13-36 cytochrome P450, family 2, subfamily d, polypeptide 1 Rattus norvegicus 70-76 32134850-9 2020 CONCLUSIONS: The combined CYP2D6/CYP2C19 phenotype has a significant impact on serum concentrations of venlafaxine and also on the active moiety of venlafaxine + ODV, than CYP2D6 alone. Desvenlafaxine Succinate 162-165 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 26-32 32134850-9 2020 CONCLUSIONS: The combined CYP2D6/CYP2C19 phenotype has a significant impact on serum concentrations of venlafaxine and also on the active moiety of venlafaxine + ODV, than CYP2D6 alone. Desvenlafaxine Succinate 162-165 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 33-40 32134850-9 2020 CONCLUSIONS: The combined CYP2D6/CYP2C19 phenotype has a significant impact on serum concentrations of venlafaxine and also on the active moiety of venlafaxine + ODV, than CYP2D6 alone. Desvenlafaxine Succinate 162-165 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 172-178