PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 34560933-1 2021 In capillary electrophoresis an enantioseparation of daclatasvir (DCV) was observed in case of heptakis(2,6-di-O-methyl)-beta-CD, heptakis(2-O-methyl)-beta-CD and beta-CD, while two peaks with a plateau were noted for heptakis(2,3,6-tri-O-methyl)-beta-CD and heptakis(2,3-di-O-methyl)-beta-CD indicating a slow equilibrium. daclatasvir 53-64 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 121-128 34560933-1 2021 In capillary electrophoresis an enantioseparation of daclatasvir (DCV) was observed in case of heptakis(2,6-di-O-methyl)-beta-CD, heptakis(2-O-methyl)-beta-CD and beta-CD, while two peaks with a plateau were noted for heptakis(2,3,6-tri-O-methyl)-beta-CD and heptakis(2,3-di-O-methyl)-beta-CD indicating a slow equilibrium. daclatasvir 53-64 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 151-158 34560933-1 2021 In capillary electrophoresis an enantioseparation of daclatasvir (DCV) was observed in case of heptakis(2,6-di-O-methyl)-beta-CD, heptakis(2-O-methyl)-beta-CD and beta-CD, while two peaks with a plateau were noted for heptakis(2,3,6-tri-O-methyl)-beta-CD and heptakis(2,3-di-O-methyl)-beta-CD indicating a slow equilibrium. daclatasvir 53-64 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 163-170 34560933-1 2021 In capillary electrophoresis an enantioseparation of daclatasvir (DCV) was observed in case of heptakis(2,6-di-O-methyl)-beta-CD, heptakis(2-O-methyl)-beta-CD and beta-CD, while two peaks with a plateau were noted for heptakis(2,3,6-tri-O-methyl)-beta-CD and heptakis(2,3-di-O-methyl)-beta-CD indicating a slow equilibrium. daclatasvir 53-64 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 247-254 34560933-1 2021 In capillary electrophoresis an enantioseparation of daclatasvir (DCV) was observed in case of heptakis(2,6-di-O-methyl)-beta-CD, heptakis(2-O-methyl)-beta-CD and beta-CD, while two peaks with a plateau were noted for heptakis(2,3,6-tri-O-methyl)-beta-CD and heptakis(2,3-di-O-methyl)-beta-CD indicating a slow equilibrium. daclatasvir 53-64 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 285-292 34560933-6 2021 Higher order complexes between DCV and heptakis(2,6-di-O-methyl)-beta-CD were corroborated by mass spectrometry. daclatasvir 31-34 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 65-72 31858513-1 2020 NS5A-P32 deletion (P32del) is a resistance-associated amino acid change that has recently gained popularity in direct-acting antiviral treatment for chronic hepatitis C. Although not yet detected in naive patients, it appears in 5 to 10% of hepatitis C genotype 1b patients who fail to respond to daclatasvir/asunaprevir and sofosbuvir/ledipasvir treatments. daclatasvir 297-308 inhibitor of growth family member 2 Homo sapiens 5-8 33857308-0 2021 Erratum to: SD1000: High Sustained Viral Response Rate in 1361 Patients With Hepatitis C Genotypes 1, 2, 3, and 4 Using a Low-cost Fixed-dose Combination Tablet of Generic Sofosbuvir and Daclatasvir: A Multicenter Phase III Clinical Trial. daclatasvir 187-198 hepatitis c genotypes 1, 2, 3, and 4 None 77-113 32804853-1 2021 OBJECTIVES: To evaluate the effect of generic sofosbuvir and daclatasvir (SOF/DCV) treatment on the glycemic state and insulin resistance as well as lipid profiles of those who achieved sustained virological response (SVR) in diabetic chronic hepatitis C virus (CHC) patients. daclatasvir 61-72 insulin Homo sapiens 119-126 35379125-2 2022 METHODS: The transport of daclatasvir, as well as the standard rhodamine 123 by P-gp across the rat intestine, was studied in vitro using the non-everted sac method. daclatasvir 26-37 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 80-84 35379125-6 2022 RESULTS: The inhibitory effect of atorvastatin on Pgp activity and expression was manifested by increased serosal transport of the standard rhodamine 123, as well as daclatasvir. daclatasvir 166-177 phosphoglycolate phosphatase Rattus norvegicus 50-53 35379125-11 2022 CONCLUSION: Atorvastatin has a significant effect on P-gp mediated intestinal transport of daclatasvir, however, it did not affect the systemic bioavailability of a single oral dose of daclatasvir. daclatasvir 91-102 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 53-57 34997607-10 2022 Daclatasvir showed site dependent intestinal absorption in a manner suggesting its affection by P-gp efflux. daclatasvir 0-11 ATP binding cassette subfamily B member 1 Homo sapiens 96-100 33218024-7 2020 Furthermore, three of the top 30 drugs with strong affinity prediction for the TMPRSS2 are anti-hepatitis C virus (HCV) drugs, including ombitasvir, daclatasvir, and paritaprevir. daclatasvir 149-160 transmembrane serine protease 2 Homo sapiens 79-86 32982753-14 2020 However, SOF/VEL+RBV achieved a high SVR rate in cirrhotic patients then SOF+DCV+RBV. daclatasvir 77-80 small integral membrane protein 1 (Vel blood group) Homo sapiens 9-16 32486887-0 2020 Peripheral Expression of CXCL10 Gene in Chronic Hepatitis C Patients Treated with Sofosbuvir, Daclatasvir, and Ribavirin. daclatasvir 94-105 C-X-C motif chemokine ligand 10 Homo sapiens 25-31 32486887-6 2020 In this study we analyzed the expression levels of CXCL10 mRNA in the 90 chronic HCV patients using quantitative PCR (qPCR) prior, after, and during therapy with sofosbuvir/ribavirin (SOF+RBV) and sofosbuvir/daclatasvir/ribavirin (SOF+DCV+RBV), and further, the results were analyzed relative to treatment response. daclatasvir 235-238 C-X-C motif chemokine ligand 10 Homo sapiens 51-57 32486887-10 2020 Comparing the 2 regimens, the reduction in peripheral CXCL10 expression was more pronounced in patients undergoing SOF+DCV+RBV therapy. daclatasvir 119-122 C-X-C motif chemokine ligand 10 Homo sapiens 54-60 31959479-1 2020 BACKGROUND: Daclatasvir (DCV) is a HCV NS5A inhibitor whose plasma exposure may be influenced by co-administration with inducers or inhibitors of CYP3A4 such as many antiretrovirals. daclatasvir 12-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 31863490-8 2020 We have studied the expression levels of one of these antiviral effectors, TRIM22 in response to sofosbuvir (SOF) and daclatasvir (DAC) in combination with RBV, using quantitative PCR in the peripheral blood mononuclear cells (PBMCs) of HCV-infected patients. daclatasvir 118-129 tripartite motif containing 22 Homo sapiens 75-81 31863490-8 2020 We have studied the expression levels of one of these antiviral effectors, TRIM22 in response to sofosbuvir (SOF) and daclatasvir (DAC) in combination with RBV, using quantitative PCR in the peripheral blood mononuclear cells (PBMCs) of HCV-infected patients. daclatasvir 131-134 tripartite motif containing 22 Homo sapiens 75-81 31959479-1 2020 BACKGROUND: Daclatasvir (DCV) is a HCV NS5A inhibitor whose plasma exposure may be influenced by co-administration with inducers or inhibitors of CYP3A4 such as many antiretrovirals. daclatasvir 25-28 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 146-152 31914336-5 2020 Moreover, DCV-TRIO lacks differentiation from its earlier-launched DAA rivals, presents an inconvenient twice-daily dosing schedule and is not recommended in patients with advanced liver and kidney disease. daclatasvir 10-13 trio Rho guanine nucleotide exchange factor Homo sapiens 14-18 31846553-7 2020 RESULTS: Twenty of 43 (46.5%) daclatasvir+asunaprevir (DCV+ASV)-treated patients with virologic failure had no RASs at baseline, 3 (15%) acquired P32 deletion RASs. daclatasvir 30-41 inhibitor of growth family member 2 Homo sapiens 146-149 32012839-5 2020 Compared with "Treat at F3"; "Treat at F0" exhibited an ICER of US$3780.20/QALY and US$15,145.98/QALY under the DCV+ASV regimen and PegIFN-based regimen; respectively. daclatasvir 112-115 cAMP responsive element modulator Homo sapiens 56-60 31527804-9 2019 However, the hsa-miR146b-5p expression in CD14+ monocytes of SVR patients treated with DCV and ASV was comparable to that in monocytes of non-SVR patients treated with DCV and ASV. daclatasvir 87-90 CD14 molecule Homo sapiens 42-46 31951178-8 2020 RESULTS: SOF and DAC exerted a potent antifibrotic effect evidenced by their activity against hyaluronic acid HA and metalloproteinase MMP-9 significantly (P<=0.001). daclatasvir 17-20 matrix metallopeptidase 9 Homo sapiens 135-140 31645655-0 2020 Effect of CYP3A5*3 genetic variant on the metabolism of direct-acting antivirals in vitro: a different effect on asunaprevir versus daclatasvir and beclabuvir. daclatasvir 132-143 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 10-16 31645655-1 2020 Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. daclatasvir 45-56 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 203-209 31645655-1 2020 Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. daclatasvir 45-56 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 214-220 31645655-1 2020 Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. daclatasvir 58-61 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 203-209 31645655-1 2020 Direct-acting antivirals, asunaprevir (ASV), daclatasvir (DCV), and beclabuvir (BCV) are known to be mainly metabolized by CYP3A enzymes; however, the differences in the detailed metabolic activities of CYP3A4 and CYP3A5 on these drugs are not well clarified. daclatasvir 58-61 cytochrome P450 family 3 subfamily A member 5 Homo sapiens 214-220 31645655-2 2020 The aim of the present study was to elucidate the relative contributions of CYP3A4 and CYP3A5 to the metabolism of ASV, DCV, and BCV, as well as the effect of CYP3A5*3 genetic variant in vitro. daclatasvir 120-123 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 76-82 31914336-3 2020 Currently, several DAA options are available on the market.Areas covered: This review focuses on the pharmacokinetics, efficacy, tolerability and safety profile of DCV-TRIO, a twice-daily fixed-dose combination of daclatasvir, asunaprevir and beclabuvir approved in Japan for the treatment of genotype 1 HCV infection.Expert opinion: The DCV-TRIO combination achieved good response rates in genotype 1 patients (SVR12 >= 95% in naive subtype 1b), independently from IL28B genotype, cirrhotic status and prior interferon exposure. daclatasvir 164-167 trio Rho guanine nucleotide exchange factor Homo sapiens 168-172 31914336-3 2020 Currently, several DAA options are available on the market.Areas covered: This review focuses on the pharmacokinetics, efficacy, tolerability and safety profile of DCV-TRIO, a twice-daily fixed-dose combination of daclatasvir, asunaprevir and beclabuvir approved in Japan for the treatment of genotype 1 HCV infection.Expert opinion: The DCV-TRIO combination achieved good response rates in genotype 1 patients (SVR12 >= 95% in naive subtype 1b), independently from IL28B genotype, cirrhotic status and prior interferon exposure. daclatasvir 164-167 trio Rho guanine nucleotide exchange factor Homo sapiens 342-346 31914336-3 2020 Currently, several DAA options are available on the market.Areas covered: This review focuses on the pharmacokinetics, efficacy, tolerability and safety profile of DCV-TRIO, a twice-daily fixed-dose combination of daclatasvir, asunaprevir and beclabuvir approved in Japan for the treatment of genotype 1 HCV infection.Expert opinion: The DCV-TRIO combination achieved good response rates in genotype 1 patients (SVR12 >= 95% in naive subtype 1b), independently from IL28B genotype, cirrhotic status and prior interferon exposure. daclatasvir 164-167 interferon lambda 3 Homo sapiens 466-471 32367815-10 2020 CONCLUSIONS: Body weight and serum albumin were the major determinants of daclatasvir V/F in this population. daclatasvir 74-85 albumin Homo sapiens 35-42 31481446-4 2019 Lopinavir, ritonavir, saquinavir, atazanavir, maraviroc, ledipasvir, and daclatasvir inhibited the efflux of a model ABCB1 substrate, rhodamine 123 (RHD123), in Caco-2 cells and rat-derived PCIS. daclatasvir 73-84 ATP binding cassette subfamily B member 1 Homo sapiens 117-122 31380801-3 2019 However, DCV[3] and DCV[4] crystallized in the centrosymmetric space group P21/c which excludes their application as nonlinear optical materials in the crystalline state. daclatasvir 20-23 H3 histone pseudogene 16 Homo sapiens 75-78 30848363-9 2019 CONCLUSIONS: Many patients treated with the DCV/ASV/BCV regimen have a history of a failure to achieve SVR with previous IFN-free DAA therapy. daclatasvir 44-47 interferon alpha 1 Homo sapiens 121-124 30848363-11 2019 In addition, most patients with a history of failure with IFN-free DAA therapy, particularly the DCV/ASV regimen, showed resistance to this regimen. daclatasvir 97-100 interferon alpha 1 Homo sapiens 58-61 29094205-0 2018 Association between alanine aminotransferase elevation and UGT1A1*6 polymorphisms in daclatasvir and asunaprevir combination therapy for chronic hepatitis C. BACKGROUND: Liver damage presented as alanine aminotransferase (ALT) elevation and high ALT-caused treatment discontinuation occurs with high frequency in Japanese patients receiving daclatasvir plus asunaprevir (DCV/ASV) therapy for hepatitis C virus (HCV) infection, and its mechanism is unknown. daclatasvir 85-96 glutamic--pyruvic transaminase Homo sapiens 20-44 31291311-0 2019 Single nucleotide polymorphisms associated with elevated alanine aminotransferase in patients receiving asunaprevir plus daclatasvir combination therapy for chronic hepatitis C. AIMS: Drug-induced liver damage characterized by serum alanine aminotransferase (ALT) elevation often occurs in direct-acting antiviral (DAA) combination therapy for chronic hepatitis C virus (HCV) infection. daclatasvir 121-132 glutamic--pyruvic transaminase Homo sapiens 57-81 31291311-9 2019 CONCLUSIONS: CYP3A4 rs4646437 was found to be significantly and independently associated with ALT elevation in Japanese patients receiving ASV plus DCV therapy. daclatasvir 148-151 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 13-19 30467710-4 2019 In chronic experiments, we found that nicotine and PACAP both reduced DCV and DC diameters and that this effect was likewise prevented by CST. daclatasvir 70-73 adenylate cyclase activating polypeptide 1 Rattus norvegicus 51-56 30467710-9 2019 We conclude that CST regulates DCV quanta by acutely inhibiting catecholamine secretion and chronically increasing expression of CgA after nicotinic stimulation and CgB and SgII after PACAPergic stimulation. daclatasvir 31-34 chromogranin A Rattus norvegicus 129-132 30906544-0 2019 Changes in serum LDL, PCSK9 and microRNA-122 in patients with chronic HCV infection receiving Daclatasvir/Asunaprevir. daclatasvir 94-105 proprotein convertase subtilisin/kexin type 9 Homo sapiens 22-27 30906544-10 2019 In summary, treatment of HCV with Daclatasvir/Asunaprevir resulted in elevated LDL, and relative miR122 and PCSK9-A levels in serum appeared to have some association with LDL increase. daclatasvir 34-45 microRNA 122 Homo sapiens 97-103 30906544-10 2019 In summary, treatment of HCV with Daclatasvir/Asunaprevir resulted in elevated LDL, and relative miR122 and PCSK9-A levels in serum appeared to have some association with LDL increase. daclatasvir 34-45 proprotein convertase subtilisin/kexin type 9 Homo sapiens 108-113 30266283-0 2019 Real-world effectiveness of daclatasvir plus sofosbuvir and velpatasvir/sofosbuvir in hepatitis C genotype 2 and 3. daclatasvir 28-39 c genotype 2 and 3 None 96-114 30067145-0 2018 Decreased CD4+CD25+CD127dim/- Regulatory T Cells and T Helper 17 Cell Responsiveness to Toll-Like Receptor 2 in Chronic Hepatitis C Patients with Daclatasvir Plus Asunaprevir Therapy. daclatasvir 146-157 CD4 molecule Homo sapiens 10-13 30067145-11 2018 These data suggested that daclatasvir plus asunaprevir therapy resulted in the decreased responsiveness of Tregs/Th17 cells to TLR2 stimulation in chronic hepatitis C patients, which might provide a novel mechanism underlying DAA-induced immunoregulation. daclatasvir 26-37 toll like receptor 2 Homo sapiens 127-131 29864695-5 2018 The linear ranges were 1-20 mug mL-1 for SOF and 0.6-6 mug mL-1 for DCS with correlation coefficients >=0.9995. daclatasvir 68-71 L1 cell adhesion molecule Mus musculus 32-54 29500489-0 2018 Potent viral suppression and improvements in alpha-fetoprotein and measures of fibrosis in Japanese patients receiving a daclatasvir/asunaprevir/beclabuvir fixed-dose combination for the treatment of HCV genotype-1 infection. daclatasvir 121-132 alpha fetoprotein Homo sapiens 45-62 29500489-3 2018 METHODS: Treatment-naive or interferon-experienced UNITY-3 patients with HCV GT-1 who received twice-daily DCV-TRIO were assessed for fibrosis [FibroTest; FibroScan; fibrosis-4 index (FIB-4), aspartate-aminotransferase-to-platelet-ratio index] and AFP at baseline and Weeks 4 (FIB-4 only), 12 or 24 post-treatment. daclatasvir 107-110 trio Rho guanine nucleotide exchange factor Homo sapiens 111-115 29500489-8 2018 CONCLUSIONS: DCV-TRIO administered for 12 weeks to Japanese patients with primarily GT-1b infection achieved a high SVR12 rate and resulted in improved measures of hepatic fibrosis and serum AFP that may reduce the risk of future liver disease progression and hepatocellular carcinoma, particularly in those with compensated cirrhosis. daclatasvir 13-16 trio Rho guanine nucleotide exchange factor Homo sapiens 17-21 29500489-8 2018 CONCLUSIONS: DCV-TRIO administered for 12 weeks to Japanese patients with primarily GT-1b infection achieved a high SVR12 rate and resulted in improved measures of hepatic fibrosis and serum AFP that may reduce the risk of future liver disease progression and hepatocellular carcinoma, particularly in those with compensated cirrhosis. daclatasvir 13-16 alpha fetoprotein Homo sapiens 191-194 29353349-8 2018 Daclatasvir has low-to-moderate clearance with the predominant route of elimination via cytochrome P450 3A4-mediated metabolism and P-glycoprotein excretion and intestinal secretion. daclatasvir 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 88-107 29353349-10 2018 As a result, the dose of daclatasvir should be reduced from 60 to 30 mg once daily when co-administered with strong inhibitors of cytochrome P450 3A4. daclatasvir 25-36 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 130-149 29353349-12 2018 The dose of daclatasvir should be increased from 60 to 90 mg once daily when co-administered with moderate inducers of cytochrome P450 3A4. daclatasvir 12-23 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 119-138 29353349-13 2018 Co-administration of daclatasvir with strong inducers of cytochrome P450 3A4 is contraindicated. daclatasvir 21-32 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 57-76 29353349-16 2018 In vitro, daclatasvir is a weak-to-moderate inhibitor of transporters including organic cation transporter 1, P-glycoprotein, organic transporting polypeptide 1B1, organic transporting polypeptide 1B3, and breast cancer resistance protein, although in clinical studies, daclatasvir has not altered the pharmacokinetics of concomitantly administered drugs that are substrates of these transporters to an appreciable degree, except for rosuvastatin. daclatasvir 10-21 solute carrier family 22 member 1 Homo sapiens 80-108 29353349-16 2018 In vitro, daclatasvir is a weak-to-moderate inhibitor of transporters including organic cation transporter 1, P-glycoprotein, organic transporting polypeptide 1B1, organic transporting polypeptide 1B3, and breast cancer resistance protein, although in clinical studies, daclatasvir has not altered the pharmacokinetics of concomitantly administered drugs that are substrates of these transporters to an appreciable degree, except for rosuvastatin. daclatasvir 10-21 ATP binding cassette subfamily B member 1 Homo sapiens 110-200 29855221-1 2018 Areas covered: This article reviews the pharmacodynamic and pharmacokinetic properties of the DCV-TRIO combination. daclatasvir 94-97 trio Rho guanine nucleotide exchange factor Homo sapiens 98-102 29454794-9 2018 Compared with an HCV genotype 1a recombinant, HCV genotype 6a recombinants of strains HK2 and HK6a were equally sensitive to daclatasvir, elbasvir, velpatasvir, pibrentasvir, and sofosbuvir, but less sensitive to ledipasvir, ombitasvir, and dasabuvir. daclatasvir 125-136 hexokinase 2 Homo sapiens 86-89 30431653-0 2019 Polymorphism in interferon lambda3/interleukin-28B gene and risk to noncirrhotic chronic hepatitis C genotype 3 virus infection and its effect on the response to combined daclatasvir and sofosbuvir therapy. daclatasvir 171-182 interferon lambda 3 Homo sapiens 16-34 30431653-0 2019 Polymorphism in interferon lambda3/interleukin-28B gene and risk to noncirrhotic chronic hepatitis C genotype 3 virus infection and its effect on the response to combined daclatasvir and sofosbuvir therapy. daclatasvir 171-182 interferon lambda 3 Homo sapiens 35-50 30431653-2 2019 The present study was, therefore, initiated to investigate the association of polymorphism in interferon lambda3 (IFNL3) also known as interleukin-28B (IL-28B) gene with chronic HCV infection and association of these polymorphic variants with the combination daclatasvir and sofosbuvir HCV therapy response. daclatasvir 259-270 interferon lambda 3 Homo sapiens 94-112 30431653-2 2019 The present study was, therefore, initiated to investigate the association of polymorphism in interferon lambda3 (IFNL3) also known as interleukin-28B (IL-28B) gene with chronic HCV infection and association of these polymorphic variants with the combination daclatasvir and sofosbuvir HCV therapy response. daclatasvir 259-270 interferon lambda 3 Homo sapiens 114-119 30431653-2 2019 The present study was, therefore, initiated to investigate the association of polymorphism in interferon lambda3 (IFNL3) also known as interleukin-28B (IL-28B) gene with chronic HCV infection and association of these polymorphic variants with the combination daclatasvir and sofosbuvir HCV therapy response. daclatasvir 259-270 interferon lambda 3 Homo sapiens 135-150 30431653-2 2019 The present study was, therefore, initiated to investigate the association of polymorphism in interferon lambda3 (IFNL3) also known as interleukin-28B (IL-28B) gene with chronic HCV infection and association of these polymorphic variants with the combination daclatasvir and sofosbuvir HCV therapy response. daclatasvir 259-270 interferon lambda 3 Homo sapiens 152-158 30253037-0 2018 Association of IL-1beta, IL-1RN, and ESR1 genes polymorphism with risk of infection and response to sofosbuvir plus daclatasvir combination therapy in hepatitis C virus genotype-4 patients. daclatasvir 116-127 estrogen receptor 1 Homo sapiens 37-41 30344416-6 2018 P32 deletion, an NS5A RAS, has been gradually noticed in patients with DCV/ASV failure. daclatasvir 71-74 inhibitor of growth family member 2 Homo sapiens 0-3 29094205-0 2018 Association between alanine aminotransferase elevation and UGT1A1*6 polymorphisms in daclatasvir and asunaprevir combination therapy for chronic hepatitis C. BACKGROUND: Liver damage presented as alanine aminotransferase (ALT) elevation and high ALT-caused treatment discontinuation occurs with high frequency in Japanese patients receiving daclatasvir plus asunaprevir (DCV/ASV) therapy for hepatitis C virus (HCV) infection, and its mechanism is unknown. daclatasvir 85-96 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 59-65 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. daclatasvir 200-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 66-72 29094205-5 2018 CONCLUSIONS: Through the analysis we suggest that the A allele in UGT1A1 rs4148323 (UGT1A1*6), which is highly prevalent in the Japanese population, should be considered a risk for the development of DCV/ASV therapy-induced ALT elevation. daclatasvir 200-203 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 84-90 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. daclatasvir 103-106 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29094205-6 2018 Pretreatment SNP testing of UGT1A1*6 might be beneficial for the prediction of liver damage induced by DCV/ASV or even by DCV/ASV plus beclabuvir. daclatasvir 122-125 UDP glucuronosyltransferase family 1 member A1 Homo sapiens 28-34 29484572-11 2018 CONCLUSION: Low-dose Sofosbuvir and full-dose Daclatasvir are safe and effective in treating CHC in patients with CKD with eGFR less than 30 mL/min/1.73 m2. daclatasvir 46-57 CD59 molecule (CD59 blood group) Homo sapiens 144-149 29790402-7 2018 DCV levels were influenced by CYP24A1 rs2248359T>C polymorphism at 2 weeks and VDR Cdx2 A>G at 1 month of treatment. daclatasvir 0-3 cytochrome P450 family 24 subfamily A member 1 Homo sapiens 30-37 29790402-7 2018 DCV levels were influenced by CYP24A1 rs2248359T>C polymorphism at 2 weeks and VDR Cdx2 A>G at 1 month of treatment. daclatasvir 0-3 vitamin D receptor Homo sapiens 82-85 29790402-7 2018 DCV levels were influenced by CYP24A1 rs2248359T>C polymorphism at 2 weeks and VDR Cdx2 A>G at 1 month of treatment. daclatasvir 0-3 caudal type homeobox 2 Homo sapiens 86-90 28892235-11 2018 In conclusion, this analysis showed high response rates in HCV genotype 1-infected patients treated with daclatasvir, ASV, and BCV irrespective of RBV use, prior interferon-based therapy, or restriction on non-cirrhotic patients, IL28B genotype, or baseline resistance-associated variants. daclatasvir 105-116 interferon lambda 3 Homo sapiens 230-235 29327780-8 2018 The pooled SVR rates for GT1 were 89.9% (95% CI 88.6-91.1, I2 = 55.1%) with daclatasvir/asunaprevir (DCV/ASV) and 98.1% (95% CI 97.0-99.0, I2 = 41.0%) with ledipasvir/sofosbuvir +- ribavirin (LDV/SOF +- RBV). daclatasvir 77-88 beta-1,4-galactosyltransferase 1 Homo sapiens 25-28 29414007-7 2018 Under the optimum conditions, the linear ranges of calibration curves were 0.2-30 and 6-120 ng mL-1 for DCS and LDS, respectively with correlation coefficients >=0.9998. daclatasvir 104-107 L1 cell adhesion molecule Mus musculus 95-99 29414007-8 2018 The detection limits were 0.047 and 1.939 ng mL-1 for DCS and LDS, respectively indicating ultrasensitivity of the proposed method. daclatasvir 54-57 L1 cell adhesion molecule Mus musculus 45-49 29869645-1 2018 The fixed-dose tablet combination of daclatasvir (30 mg)/asunaprevir (200 mg)/beclabuvir (75 mg), DCV-TRIO, is one of the latest drugs in the pipeline of interferon-free direct-acting antiviral hepatitis C virus (HCV) therapies. daclatasvir 37-48 trio Rho guanine nucleotide exchange factor Homo sapiens 102-106 29869645-1 2018 The fixed-dose tablet combination of daclatasvir (30 mg)/asunaprevir (200 mg)/beclabuvir (75 mg), DCV-TRIO, is one of the latest drugs in the pipeline of interferon-free direct-acting antiviral hepatitis C virus (HCV) therapies. daclatasvir 98-101 trio Rho guanine nucleotide exchange factor Homo sapiens 102-106 29869645-3 2018 Results from the UNITY 1, 2, 3 and 4 phase III clinical trials showed that DCV-TRIO exhibited high sustained virologic responses at 12 weeks (between 92% and 100% for HCV GT-1 treatment-naive patients). daclatasvir 75-78 trio Rho guanine nucleotide exchange factor Homo sapiens 79-83 29869645-5 2018 Further research should focus on more real-life data on DCV-TRIO and on developing a pill regimen that works on other HCV genotypes with high genetic barriers and that is available at a reduced cost. daclatasvir 56-59 trio Rho guanine nucleotide exchange factor Homo sapiens 60-64 28815329-1 2018 BACKGROUND: In Japan, daclatasvir (DCV) and asunaprevir (ASV) therapy was the first IFN-free treatment to be approved, and thousands of patients have since been successfully treated, with an SVR rate of around 90%. daclatasvir 22-33 interferon alpha 1 Homo sapiens 84-87 28815329-1 2018 BACKGROUND: In Japan, daclatasvir (DCV) and asunaprevir (ASV) therapy was the first IFN-free treatment to be approved, and thousands of patients have since been successfully treated, with an SVR rate of around 90%. daclatasvir 35-38 interferon alpha 1 Homo sapiens 84-87 29327780-8 2018 The pooled SVR rates for GT1 were 89.9% (95% CI 88.6-91.1, I2 = 55.1%) with daclatasvir/asunaprevir (DCV/ASV) and 98.1% (95% CI 97.0-99.0, I2 = 41.0%) with ledipasvir/sofosbuvir +- ribavirin (LDV/SOF +- RBV). daclatasvir 102-105 beta-1,4-galactosyltransferase 1 Homo sapiens 25-28 28884930-5 2018 Among the 6 patients receiving lead-in injections, viral relapse occurred in 2 patients who had an unfavorable IFN-lambda3-related gene single nucleotide polymorphism allele; both patients had been previously treated with simeprevir, and HCV carrying NS5A-L31V/Y93H mutations had emerged after DCV/ASV. daclatasvir 294-297 interferon lambda 3 Homo sapiens 111-122 29255971-0 2018 Effects of a Fixed-Dose Co-Formulation of Daclatasvir, Asunaprevir, and Beclabuvir on the Pharmacokinetics of a Cocktail of Cytochrome P450 and Drug Transporter Substrates in Healthy Subjects. daclatasvir 42-53 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 124-139 28834042-7 2018 RESULTS: In 69 (81.2%) patients who received DCV/ASV and achieved a sustained virologic response at 24 weeks after the end of treatment (SVR24), TC and LDL-C increased significantly from baseline to p24w. daclatasvir 45-48 component of oligomeric golgi complex 2 Homo sapiens 152-157 29743792-14 2018 Further wider-scale studies are needed to evaluate the actual role of IL18 polymorphisms in treatment response with sofosbuvir/daclatasvir. daclatasvir 127-138 interleukin 18 Homo sapiens 70-74 29111569-8 2018 In contrast, addition of either mTOR inhibitor to DAC induced a 30% reduction in antiviral activity in GT1b cells (P <= .01 vs DAC alone). daclatasvir 50-53 mechanistic target of rapamycin kinase Homo sapiens 32-36 29111569-8 2018 In contrast, addition of either mTOR inhibitor to DAC induced a 30% reduction in antiviral activity in GT1b cells (P <= .01 vs DAC alone). daclatasvir 130-133 mechanistic target of rapamycin kinase Homo sapiens 32-36 29091211-0 2017 Influence of ABCB11 and HNF4alpha genes on daclatasvir plasma concentration: preliminary pharmacogenetic data from the Kineti-C study. daclatasvir 43-54 hepatocyte nuclear factor 4 alpha Homo sapiens 24-33 28834042-10 2018 At 12w, TC and LDL-C increased to a greater degree in patients receiving SOF/LDV than in those receiving DCV/ASV or SOF/RBV. daclatasvir 105-108 component of oligomeric golgi complex 2 Homo sapiens 15-20 29111569-6 2018 RESULTS: Addition of either mTOR inhibitor to the DAA daclatasvir (DAC) resulted in a 30% increase in antiviral activity compared to DAC alone for HCV GT2a, GT3a, and GT4a (all P <= .01). daclatasvir 67-70 mechanistic target of rapamycin kinase Homo sapiens 28-32 29111569-6 2018 RESULTS: Addition of either mTOR inhibitor to the DAA daclatasvir (DAC) resulted in a 30% increase in antiviral activity compared to DAC alone for HCV GT2a, GT3a, and GT4a (all P <= .01). daclatasvir 133-136 mechanistic target of rapamycin kinase Homo sapiens 28-32 28370350-11 2017 CONCLUSIONS: Twelve weeks of DCV-TRIO was well tolerated and provided 100% SVR12 in treatment-naive and treatment-experienced patients with genotype-1 infection, with or without cirrhosis, including those with baseline NS5A-L31 or NS5A-Y93 resistance-associated substitutions. daclatasvir 29-32 trio Rho guanine nucleotide exchange factor Homo sapiens 33-37 29091211-3 2017 Objectives: We investigated the association between daclatasvir plasma concentrations at 2 weeks and 1 month of therapy and genetic variants (SNPs) in genes encoding transporters and nuclear factors (ABCB1, ABCB11 and HNF4alpha). daclatasvir 52-63 ATP binding cassette subfamily B member 1 Homo sapiens 200-205 29091211-3 2017 Objectives: We investigated the association between daclatasvir plasma concentrations at 2 weeks and 1 month of therapy and genetic variants (SNPs) in genes encoding transporters and nuclear factors (ABCB1, ABCB11 and HNF4alpha). daclatasvir 52-63 ATP binding cassette subfamily B member 11 Homo sapiens 207-213 29091211-3 2017 Objectives: We investigated the association between daclatasvir plasma concentrations at 2 weeks and 1 month of therapy and genetic variants (SNPs) in genes encoding transporters and nuclear factors (ABCB1, ABCB11 and HNF4alpha). daclatasvir 52-63 hepatocyte nuclear factor 4 alpha Homo sapiens 218-227 29091211-6 2017 HNF4alpha 975 C > G polymorphism was found to be associated with the daclatasvir plasma concentrations at 2 weeks (P = 0.009) and 1 month of therapy (P = 0.006). daclatasvir 72-83 hepatocyte nuclear factor 4 alpha Homo sapiens 0-9 29085787-0 2017 Increase in Albumin by Daclatasvir/asunaprevir Therapy is Correlated with Decrease in Aspartate Transaminase. daclatasvir 23-34 solute carrier family 17 member 5 Homo sapiens 86-108 29085787-8 2017 CONCLUSION: DCV/ASV therapy resulted in an increase in albumin levels in SVR patients, which was significantly correlated with a decrease in AST levels. daclatasvir 12-15 solute carrier family 17 member 5 Homo sapiens 141-144 28797106-9 2017 Serum AFP levels decreased to similar levels between PEG-IFN/RBV and DCV/ASV groups following the achievement of viral eradication. daclatasvir 69-72 alpha fetoprotein Homo sapiens 6-9 27943563-4 2017 Patients without SVR12 were offered retreatment based on the DAA resistance profile at failure; patients with resistance to <=1 DCV-TRIO component received DCV-TRIO + RBV for 12 weeks. daclatasvir 131-134 trio Rho guanine nucleotide exchange factor Homo sapiens 135-139 27943563-9 2017 All 15 patients retreated with DCV-TRIO + RBV for 12 weeks achieved SVR12. daclatasvir 31-34 trio Rho guanine nucleotide exchange factor Homo sapiens 35-39 27943563-12 2017 Non-SVR12 was not associated with emergence of NS3 or NS5B RAS and retreatment with DCV-TRIO + RBV for 12 weeks led to SVR in all patients. daclatasvir 84-87 trio Rho guanine nucleotide exchange factor Homo sapiens 88-92 28380010-1 2017 The DNA intercalating dye Hoechst 33342 or its close analog DCV are actively removed from cells by the multidrug resistance transporter ABCG2, a protein overexpressed in metastatic cells and somatic stem cells. daclatasvir 60-63 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 136-141 26733671-7 2017 Successful QUAD therapy as well as successful IFN-free DCV/ASV regimen resulted in an improved functional NK cell response (degranulation and TRAIL expression) to in vitro stimulation with IFNalpha. daclatasvir 55-58 interferon alpha 1 Homo sapiens 46-49 26733671-7 2017 Successful QUAD therapy as well as successful IFN-free DCV/ASV regimen resulted in an improved functional NK cell response (degranulation and TRAIL expression) to in vitro stimulation with IFNalpha. daclatasvir 55-58 TNF superfamily member 10 Homo sapiens 142-147 26733671-7 2017 Successful QUAD therapy as well as successful IFN-free DCV/ASV regimen resulted in an improved functional NK cell response (degranulation and TRAIL expression) to in vitro stimulation with IFNalpha. daclatasvir 55-58 interferon alpha 1 Homo sapiens 189-197 27932311-7 2017 RESULTS: Short exposure of HCV-infected cells to daclatasvir reduced viral assembly and induced clustering of structural proteins with non-structural HCV proteins, including core, E2, NS4B, and NS5A. daclatasvir 49-60 polyprotein;protein F Hepatitis C virus genotype 1 184-188 27502287-2 2017 METHODS: In this phase 3 study, DCV-TRIO for 12 weeks and daclatasvir plus asunaprevir (DUAL) for 24 weeks were studied in Japanese patients infected with HCV genotype 1 (99 % genotype 1b). daclatasvir 32-35 trio Rho guanine nucleotide exchange factor Homo sapiens 36-40 27502287-3 2017 RESULTS: SVR12 rates >=95 % were achieved in both treatment-naive (N = 152) and interferon-experienced (N = 65) cohorts treated with DCV-TRIO for 12 weeks and were comparable across patient subgroups, including patients aged >=65 years and those with cirrhosis. daclatasvir 136-139 trio Rho guanine nucleotide exchange factor Homo sapiens 140-144 27502287-5 2017 In the absence of baseline resistance-associated polymorphisms at positions NS5A-Y93H or -L31, SVR12 rates were 98 % with DCV-TRIO or DUAL. daclatasvir 122-125 trio Rho guanine nucleotide exchange factor Homo sapiens 126-130 27502287-6 2017 Among genotype 1b-infected patients with baseline Y93H or L31 polymorphisms, 35/38 (92 %) DCV-TRIO recipients, and 7/16 (44 %) DUAL recipients achieved SVR12. daclatasvir 90-93 trio Rho guanine nucleotide exchange factor Homo sapiens 94-98 27502287-7 2017 Adverse events, mostly liver related, led to treatment discontinuation in 10 % of DCV-TRIO recipients. daclatasvir 82-85 trio Rho guanine nucleotide exchange factor Homo sapiens 86-90 27502287-8 2017 In this group, SVR12 was achieved by 3/9 patients who discontinued before Week 4 and by 12/12 patients who completed >=4 weeks of DCV-TRIO. daclatasvir 133-136 trio Rho guanine nucleotide exchange factor Homo sapiens 137-141 27502287-9 2017 Treatment-related serious adverse events occurred in 4 and 3 % of DCV-TRIO and DUAL recipients, respectively. daclatasvir 66-69 trio Rho guanine nucleotide exchange factor Homo sapiens 70-74 27502287-12 2017 CONCLUSION: SVR12 was achieved by 96 % of Japanese patients with HCV genotype 1 infection after 12 weeks of treatment with the DCV-TRIO regimen. daclatasvir 127-130 trio Rho guanine nucleotide exchange factor Homo sapiens 131-135 27890790-11 2017 LAY SUMMARY: IFN-free therapy with oral direct-acting antiviral drugs (daclatasvir and asunaprevir) for HCV infection showed similar tolerability and antiviral efficacy in patients aged >=80years as in younger patients (patients aged >=70 and <80years and patients aged <70years), with an SVR rate over 90% and no severe adverse effects. daclatasvir 71-82 interferon alpha 1 Homo sapiens 13-16 27530469-7 2017 DDI are more likely to occur with 3D regimen, daclatasvir, simeprevir and ledipasvir, as they are all both substrates and inhibitors of P-gp and/or CYP3A4, than with sofosbuvir. daclatasvir 46-57 ATP binding cassette subfamily B member 1 Homo sapiens 136-140 27530469-7 2017 DDI are more likely to occur with 3D regimen, daclatasvir, simeprevir and ledipasvir, as they are all both substrates and inhibitors of P-gp and/or CYP3A4, than with sofosbuvir. daclatasvir 46-57 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 148-154 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 66-91 27348464-13 2016 In scenario analysis, including GT1b patients with and without cirrhosis who were Y93H mutation-negative, the ICER of OBV/PTV/r vs DCV/ASV was below the Japanese willingness-to-pay threshold of JPY 5 million/QALY, while the ICER of SOF/LDV vs OBV/PTV/r was above this threshold; thus, OBV/PTV/r was cost-effective. daclatasvir 131-134 beta-1,4-galactosyltransferase 1 Homo sapiens 32-35 27798211-2 2017 When combined with the CYP3A4 substrate daclatasvir, the daclatasvir dosage should be reduced from 60 to 30 mg once daily. daclatasvir 57-68 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 23-29 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 ATP binding cassette subfamily B member 1 Homo sapiens 99-113 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 ATP binding cassette subfamily B member 1 Homo sapiens 115-119 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 218-225 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 phosphoglycolate phosphatase Homo sapiens 243-291 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 0-3 ATP binding cassette subfamily G member 2 (Junior blood group) Homo sapiens 303-335 27652142-7 2016 Subgroup analysis showed the rate of RVR and SVR24 in high-dose daclatasvir (60 mg/day) group were slightly higher than the overall results; the rate of RVR in low-dose daclatasvir (10 mg/day) group was also higher than the control group, but its SVR24 rate was similar between the two groups. daclatasvir 64-75 nuclear receptor subfamily 1 group D member 2 Homo sapiens 37-40 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 184-187 ATP binding cassette subfamily B member 1 Homo sapiens 99-113 27664109-4 2016 DCV may be the object of a DDI via the induction or inhibition of cytochrome P450 (CYP) 3A4 and/or P-glycoprotein (P-gp) by the concomitant medication, or the precipitant of a DDI via DCV-based induction/inhibition of CYP 3A4 or inhibition of P-gp, organic anion transporting polypeptide 1B1/B3, and/or breast cancer resistance protein. daclatasvir 184-187 ATP binding cassette subfamily B member 1 Homo sapiens 115-119 27652142-7 2016 Subgroup analysis showed the rate of RVR and SVR24 in high-dose daclatasvir (60 mg/day) group were slightly higher than the overall results; the rate of RVR in low-dose daclatasvir (10 mg/day) group was also higher than the control group, but its SVR24 rate was similar between the two groups. daclatasvir 169-180 nuclear receptor subfamily 1 group D member 2 Homo sapiens 153-156 27492209-8 2016 Daclatasvir and asunaprevir were stopped because this was suspected to be a side-effect of these drugs, but the patient nonetheless went on to develop severe thrombocytopenia (platelet count 17,000/muL), which needed transfusions. daclatasvir 0-11 tripartite motif containing 37 Homo sapiens 198-201 27484655-7 2016 A similar reduction in EC50 in daclatasvir-resistant mutants was achieved by combining daclatasvir with anti-miR-122. daclatasvir 31-42 microRNA 122 Homo sapiens 109-116 27388424-12 2016 CONCLUSIONS: Herein, we have reported the outcomes of HCV-associated OLP in patients who received successful treatment with IFN-free DAAs, using the DCV/ASV combination therapy. daclatasvir 149-152 interferon alpha 1 Homo sapiens 124-127 26574180-10 2016 CONCLUSION: Successful viral eradication by IFN-free daclatasvir and asunaprevir therapy could lead to improved liver function parameters and reduced liver fibrosis markers and AFP levels. daclatasvir 53-64 interferon alpha 1 Homo sapiens 44-47 26574180-10 2016 CONCLUSION: Successful viral eradication by IFN-free daclatasvir and asunaprevir therapy could lead to improved liver function parameters and reduced liver fibrosis markers and AFP levels. daclatasvir 53-64 alpha fetoprotein Homo sapiens 177-180 27029743-4 2016 Cytochrome P450s were the main enzymes involved in the metabolism of daclatasvir. daclatasvir 69-80 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 0-15 26018228-7 2016 CONCLUSION: DCV staining may be used to discriminate subsets of CD34+ cells similarly to other methods which have previously defined different functional properties that can be related to the characterization, resolution, and purification of primitive hematopoietic stem cells in combination with specific useful markers for multicolor flow cytometric measurements. daclatasvir 12-15 CD34 molecule Homo sapiens 64-68 26572539-5 2016 Quantification of NE and EPI-storing dense core (DC) vesicles (DCV) revealed decreased DCV numbers in chromaffin cells in Chga-KO mice. daclatasvir 63-66 chromogranin A Mus musculus 122-126 26572539-6 2016 For both cell types, the DCV diameter in Chga-KO mice was less (100-200 nm) than in WT mice (200-350 nm). daclatasvir 25-28 chromogranin A Mus musculus 41-45 26572539-8 2016 Chga-KO mice showed an ~47 % increase in DCV/DC ratio, implying vesicle swelling due to increased osmotically active free catecholamines. daclatasvir 41-44 chromogranin A Mus musculus 0-4 26292191-0 2016 Relationships between serum asunaprevir concentration and alanine aminotransferase elevation during daclatasvir plus asunaprevir for chronic HCV genotype 1b infection. daclatasvir 100-111 glutamic--pyruvic transaminase Homo sapiens 58-82 26292191-1 2016 Alanine aminotransferase (ALT) elevations were the most frequent adverse events during all-oral combinations with daclatasvir and asunaprevir for patients with hepatitis C virus (HCV) infection, but the underline mechanisms are unclear. daclatasvir 114-125 glutamic--pyruvic transaminase Homo sapiens 0-24 26760453-13 2016 CONCLUSION: IFN- and RBV-free treatment with SOF/DCV was well tolerated and achieved SVR12 in all HIV/HCV with advanced liver disease. daclatasvir 49-52 interferon alpha 1 Homo sapiens 12-16 27022224-15 2016 Among both daclatasvir and telaprevir treatment groups, across GT1b- or GT1a-infected patients, lower response rates were observed in patients with IL28B non-CC and cirrhosis - factors known to affect response to pegIFN/RBV. daclatasvir 11-22 interferon lambda 3 Homo sapiens 148-153 26440999-11 2016 CONCLUSION: Cost-effectiveness conclusions are similar for patients treated in the chronic hepatitis C and CC disease stages, with DCV + ASV expected to be cost-saving versus standard of care in Japan for patients with HCV genotype 1b patients who have failed prior therapy or are IFN-ineligible/intolerant. daclatasvir 131-134 interferon alpha 1 Homo sapiens 281-284 25904097-1 2016 BACKGROUND: Resistance-associated variants (RAVs) reduce the efficacy of interferon (IFN)-free therapy with asunaprevir and daclatasvir for patients infected with hepatitis C virus (HCV) genotype 1b. daclatasvir 124-135 interferon alpha 1 Homo sapiens 73-90 27750228-1 2016 In Japan, in September 2014, IFN-free asunaprevir (ASV) and daclatasvir (DCV) became available for combination therapy. daclatasvir 73-76 interferon alpha 1 Homo sapiens 29-32 26313445-2 2015 This Phase III study (COMMAND-4; AI444-042) evaluated the efficacy and safety of daclatasvir (DCV), a pan-genotypic HCV NS5A inhibitor, with pegylated interferon-alpha2a/ribavirin (PEG-IFN/RBV) in treatment-naive patients with HCV GT4 infection. daclatasvir 81-92 interferon alpha 2 Homo sapiens 151-169 26544203-11 2015 The ICER of DCV/ASV compared to PR was US$ 16,635/QALY at a total treatment price of US$ 77,419; US$11,581 /QALY at a price of US$ 58,065; US$ 6,375/QALY at a price of US$ 38,710; and US$ 1,364 /QALY at a price of US$ 19,355. daclatasvir 12-15 cAMP responsive element modulator Homo sapiens 4-8 26744738-8 2015 Daclatasvir and asunaprevir are substrates for cytochrome P450 3A4 enzymatic pathway; thus, there is a substantial potential for drug interactions. daclatasvir 0-11 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 47-66 26711745-9 2015 The retreatment of resistant viruses that emerged using DCV monotherapy with DCV-NS5A-Syn resulted in a multilog drop and rebound in HCV similar to the initial decline and rebound observed with DCV alone on wild-type (WT) virus. daclatasvir 56-59 synemin Homo sapiens 86-89 26711745-9 2015 The retreatment of resistant viruses that emerged using DCV monotherapy with DCV-NS5A-Syn resulted in a multilog drop and rebound in HCV similar to the initial decline and rebound observed with DCV alone on wild-type (WT) virus. daclatasvir 77-80 synemin Homo sapiens 86-89 26711745-9 2015 The retreatment of resistant viruses that emerged using DCV monotherapy with DCV-NS5A-Syn resulted in a multilog drop and rebound in HCV similar to the initial decline and rebound observed with DCV alone on wild-type (WT) virus. daclatasvir 77-80 synemin Homo sapiens 86-89 26711745-10 2015 A triple combination of DCV, NS5A-Syn, and a DAA targeting the NS3 or NS5B protein cleared the cells of viruses that are highly resistant to DCV. daclatasvir 141-144 KRAS proto-oncogene, GTPase Homo sapiens 63-66 26711745-11 2015 Our data support the observation that the cooperative interaction of DCV and NS5A-Syn potentiates both the genotype coverage and resistance barrier of DCV, offering an additional DAA option for combination therapy and tools for explorations of NS5A function. daclatasvir 151-154 synemin Homo sapiens 82-85 25704315-2 2015 We herein report a case of progressive SCH with acute cellular rejection (ACR) and bacterial infection, which was successfully treated using IFN-free therapy with daclatasvir and asunaprevir. daclatasvir 163-174 interferon alpha 1 Homo sapiens 141-144 26313445-4 2015 DCV-treated patients with undetectable HCV RNA at weeks 4 and 12 (eRVR) received 24 weeks of DCV plus PEG-IFN/RBV; those without eRVR received an additional 24 weeks of PEG-IFN/RBV. daclatasvir 0-3 endogenous retrovirus group 3 member 1, envelope Homo sapiens 66-70 26313445-4 2015 DCV-treated patients with undetectable HCV RNA at weeks 4 and 12 (eRVR) received 24 weeks of DCV plus PEG-IFN/RBV; those without eRVR received an additional 24 weeks of PEG-IFN/RBV. daclatasvir 0-3 endogenous retrovirus group 3 member 1, envelope Homo sapiens 129-133 24943406-1 2015 Daclatasvir (DCV) and asunaprevir (ASV) are NS5A and NS3 protease-targeted antivirals respectively, currently under development for the treatment of chronic hepatitis C virus (HCV) infection. daclatasvir 0-11 KRAS proto-oncogene, GTPase Homo sapiens 53-56 25080450-6 2015 RESULTS: Overall, eRVR was achieved by 54.4% (80/147) of genotype 1-infected patients receiving daclatasvir 20 mg, 54.1% (79/146) receiving 60 mg versus 13.9% (10/72) receiving placebo. daclatasvir 96-107 endogenous retrovirus group 3 member 1, envelope Homo sapiens 18-22 25770426-0 2015 DIP1 plays an antiviral role against DCV infection in Drosophila melanogaster. daclatasvir 37-40 DISCO Interacting Protein 1 Drosophila melanogaster 0-4 25770426-7 2015 This study demonstrated that dip1 is a novel antiviral gene that restricts DCV replication in vitro and in vivo. daclatasvir 75-78 DISCO Interacting Protein 1 Drosophila melanogaster 29-33 24943406-1 2015 Daclatasvir (DCV) and asunaprevir (ASV) are NS5A and NS3 protease-targeted antivirals respectively, currently under development for the treatment of chronic hepatitis C virus (HCV) infection. daclatasvir 13-16 KRAS proto-oncogene, GTPase Homo sapiens 53-56 24943406-9 2015 In these patients, HCV RNA rebounded with ASV- and DCV- double resistant variants (NS3 D168A/V plus NS5A L31M and Y93H). daclatasvir 51-54 KRAS proto-oncogene, GTPase Homo sapiens 83-86 25546252-9 2015 However, DCV impairs late steps in PI4KA activation that requires NS5A expressed in the context of the HCV polyprotein. daclatasvir 9-12 phosphatidylinositol 4-kinase alpha Homo sapiens 35-40 25764674-3 2015 In September 2014, the first IFN-free DAAs, daclatasvir (DCV) and asunaprevir (ASV), were launched and the antiviral therapy for IFN intolerant/ineligible patients became to be possible. daclatasvir 44-55 interferon alpha 1 Homo sapiens 29-32 25764674-3 2015 In September 2014, the first IFN-free DAAs, daclatasvir (DCV) and asunaprevir (ASV), were launched and the antiviral therapy for IFN intolerant/ineligible patients became to be possible. daclatasvir 44-55 interferon alpha 1 Homo sapiens 129-132 25764674-3 2015 In September 2014, the first IFN-free DAAs, daclatasvir (DCV) and asunaprevir (ASV), were launched and the antiviral therapy for IFN intolerant/ineligible patients became to be possible. daclatasvir 57-60 interferon alpha 1 Homo sapiens 29-32 25529083-4 2015 A PEG-IFN/RBV-based regimen with simeprevir or daclatasvir is based on response-guided therapy and its efficacy depends on predictors of response to IFN. daclatasvir 47-58 interferon alpha 1 Homo sapiens 6-9 26549010-0 2015 Daclatasvir for the treatment of chronic hepatitis C. INTRODUCTION: Following more than 20 years of Interferon (IFN)-based treatment for hepatitis C virus (HCV), the understanding of viral life cycle led to the development of new antiviral drugs directly targeting HCV replication steps. daclatasvir 0-11 interferon alpha 1 Homo sapiens 112-115 26549010-1 2015 Daclatasvir (DCV) is a potent inhibitor of non-structural NS5A HCV protein with pangenotypic activity and low-moderate barrier to resistance suitable for IFN-free combination with other direct acting antivirals (DAAs). daclatasvir 0-11 interferon alpha 1 Homo sapiens 154-157 26549010-1 2015 Daclatasvir (DCV) is a potent inhibitor of non-structural NS5A HCV protein with pangenotypic activity and low-moderate barrier to resistance suitable for IFN-free combination with other direct acting antivirals (DAAs). daclatasvir 13-16 interferon alpha 1 Homo sapiens 154-157 26549010-4 2015 EXPERT OPINION: DCV in combination with other DAAs has provided IFN-free regimens with increased efficacy and tolerability. daclatasvir 16-19 interferon alpha 1 Homo sapiens 64-67 25529083-4 2015 A PEG-IFN/RBV-based regimen with simeprevir or daclatasvir is based on response-guided therapy and its efficacy depends on predictors of response to IFN. daclatasvir 47-58 interferon alpha 1 Homo sapiens 149-152 25529083-6 2015 In HCV-1a infected patients, the K80Q mutation in NS3 or the presence of NS5A variants at baseline are associated with poor response with simeprevir- or daclatasvir-containing regimens respectively. daclatasvir 153-164 KRAS proto-oncogene, GTPase Homo sapiens 50-53 23916491-5 2013 The results showed that Ran was upregulated in S2 cells in response to DCV infection. daclatasvir 71-74 RAN, member RAS oncogene family Homo sapiens 24-27 24612030-8 2014 Importantly, it seems that the IL28B status of the patients may influence the presence of Y93H mutations, resulting in different treatment responses to daclatasvir. daclatasvir 152-163 interferon lambda 3 Homo sapiens 31-36 25059552-2 2014 Daclatasvir combined with peginterferon alpha-2a and ribavirin in Japanese patients infected with genotype 1b HCV achieved sustained virological response (SVR) in 100% of treatment-naive patients, due to high rates of favorable IL28B allele and genotype 1b. daclatasvir 0-11 interferon lambda 3 Homo sapiens 228-233 25059552-6 2014 IFN-free regimens including daclatasvir and asunaprevir for genotype 1 null responders should be tailored to subtype, and preexisting NS5A-resistant variants should be evaluated carefully before choosing the drugs. daclatasvir 28-39 interferon alpha 1 Homo sapiens 0-4 24867984-6 2014 The results of our coincubation inhibition assays have shown that all tested DAAs could inhibit OATP1B1 functions and that SMV, ASV, and DCV (to a lesser extent), but not SFV, exhibited long-lasting preincubation inhibitory effects on OATP1B1 functions. daclatasvir 137-140 solute carrier organic anion transporter family member 1B1 Homo sapiens 235-242 23177287-3 2013 DCV as triple therapy in combination with pegylated interferon and ribavirin resulted in a high rate of early virologic response in treatment-naive patients with genotype 1 infection; as quadruple therapy in combination with asunaprevir (BMS-650032, NS3 protease inhibitor), pegylated interferon, and ribavirin, it resulted in a high rate of sustained virologic response in genotype 1 prior null responders. daclatasvir 0-3 KRAS proto-oncogene, GTPase Homo sapiens 250-253 23504694-12 2013 CONCLUSION: The treatment failure of daclatasvir and asunaprevir in HCV GT1a patients was associated with both NS5A and NS3 resistance variants in prior null responders. daclatasvir 37-48 KRAS proto-oncogene, GTPase Homo sapiens 120-123 23803447-7 2013 DCV infection activated the JAK/STAT pathway by 1 hour post incubation. daclatasvir 0-3 hopscotch Drosophila melanogaster 28-31 23803447-7 2013 DCV infection activated the JAK/STAT pathway by 1 hour post incubation. daclatasvir 0-3 Signal-transducer and activator of transcription protein at 92E Drosophila melanogaster 32-36 23803447-11 2013 CONCLUSIONS: In conclusion, the present study demonstrates that DCV infection may activate phagocytosis, JAK/STAT pathway and Imd pathway in the early host-virus interactions. daclatasvir 64-67 hopscotch Drosophila melanogaster 105-108 23803447-11 2013 CONCLUSIONS: In conclusion, the present study demonstrates that DCV infection may activate phagocytosis, JAK/STAT pathway and Imd pathway in the early host-virus interactions. daclatasvir 64-67 Signal-transducer and activator of transcription protein at 92E Drosophila melanogaster 109-113 23803447-11 2013 CONCLUSIONS: In conclusion, the present study demonstrates that DCV infection may activate phagocytosis, JAK/STAT pathway and Imd pathway in the early host-virus interactions. daclatasvir 64-67 immune deficiency Drosophila melanogaster 126-129 8344234-8 1993 Similarly, the faster velocities of DCV (V70, V80, and V90 velocities) were significantly slowed in both the VWF and non-VWF groups. daclatasvir 36-39 von Willebrand factor Homo sapiens 109-112 23358451-4 2013 In syd-2 loss of function mutants, the normal polarized localization of INS-22 neuropeptide-containing DCVs in motor neurons is disrupted, and DCVs accumulate in the cell body and dendrites. daclatasvir 103-107 Liprin-alpha Caenorhabditis elegans 3-8 23358451-4 2013 In syd-2 loss of function mutants, the normal polarized localization of INS-22 neuropeptide-containing DCVs in motor neurons is disrupted, and DCVs accumulate in the cell body and dendrites. daclatasvir 103-107 putative insulin-like peptide alpha-type 2 Caenorhabditis elegans 72-78 23358451-4 2013 In syd-2 loss of function mutants, the normal polarized localization of INS-22 neuropeptide-containing DCVs in motor neurons is disrupted, and DCVs accumulate in the cell body and dendrites. daclatasvir 143-147 Liprin-alpha Caenorhabditis elegans 3-8 23358451-5 2013 Time-lapse microscopy and kymograph analysis of mobile DCVs revealed that syd-2 mutants exhibit decreased numbers of DCVs moving in both anterograde and retrograde directions, and a corresponding increase in stationary DCVs in both axon commissures and dendrites. daclatasvir 55-59 Liprin-alpha Caenorhabditis elegans 74-79 23358451-6 2013 In addition, DCV run lengths and velocities were decreased in both axon commissures and dendrites of syd-2 mutants. daclatasvir 13-16 Liprin-alpha Caenorhabditis elegans 101-106 23358451-7 2013 This study shows that SYD-2 promotes bi-directional mobility of DCVs and identifies SYD-2 as a novel regulator of DCV trafficking and polarized distribution. daclatasvir 64-67 Liprin-alpha Caenorhabditis elegans 22-27 22706796-2 2012 The aim of this article is to report the first ever use of daclatasvir (DCV; also known as BMS-790052), a potent orally administered nonstructural 5A replication complex inhibitor, in combination with peginterferon alpha (PEG-IFNalpha) and ribavirin in an LT recipient. daclatasvir 59-70 interferon alpha 1 Homo sapiens 226-234 22706796-2 2012 The aim of this article is to report the first ever use of daclatasvir (DCV; also known as BMS-790052), a potent orally administered nonstructural 5A replication complex inhibitor, in combination with peginterferon alpha (PEG-IFNalpha) and ribavirin in an LT recipient. daclatasvir 72-75 interferon alpha 1 Homo sapiens 226-234 14764620-3 2004 POAG caused by DCV at MYOC has been termed "myocilin glaucoma". daclatasvir 15-18 myocilin Homo sapiens 22-26 14764620-3 2004 POAG caused by DCV at MYOC has been termed "myocilin glaucoma". daclatasvir 15-18 myocilin Homo sapiens 44-52 14764620-4 2004 Clinically, DCV at MYOC may manifest as a typical POAG, normal tension glaucoma, or ocular hypertension without glaucoma. daclatasvir 12-15 myocilin Homo sapiens 19-23 14764620-7 2004 DCV at MYOC cause POAG in interaction with environmental factors and DCV at other loci. daclatasvir 0-3 myocilin Homo sapiens 7-11 14764620-8 2004 Most DCV at MYOC are relatively young, and the Gln368Stop mutation is exclusively European in origin. daclatasvir 5-8 myocilin Homo sapiens 12-16 14764620-9 2004 The overall frequency of DCV at MYOC is similar among African, Caucasian and Asian probands with POAG. daclatasvir 25-28 myocilin Homo sapiens 32-36 14764620-10 2004 Because of this fact and the higher prevalence of POAG in African descendants compared with Caucasians or Asians, the overall frequency of DCV at MYOC is several-fold higher in the general population of African descendants, which is in part responsible for their higher prevalence of POAG. daclatasvir 139-142 myocilin Homo sapiens 146-150 8597299-5 1995 Quantitative analysis revealed that 4-8% of the mossy terminal profiles examined (n = 350) contained CCK-labeled DCVs, which corresponded to 0.03-0.2 labeled DCVs per 100 microns2 of neuropil. daclatasvir 113-117 cholecystokinin Rattus norvegicus 101-104 8597299-5 1995 Quantitative analysis revealed that 4-8% of the mossy terminal profiles examined (n = 350) contained CCK-labeled DCVs, which corresponded to 0.03-0.2 labeled DCVs per 100 microns2 of neuropil. daclatasvir 158-162 cholecystokinin Rattus norvegicus 101-104 12438120-2 2002 Current evidence indicates that CAPS functions selectively in DCV exocytosis by interacting with DCVs, the plasma membrane, and protein components of the fusion machinery. daclatasvir 97-101 calcyphosine Homo sapiens 32-36 8344234-8 1993 Similarly, the faster velocities of DCV (V70, V80, and V90 velocities) were significantly slowed in both the VWF and non-VWF groups. daclatasvir 36-39 von Willebrand factor Homo sapiens 121-124 33768560-0 2021 Association between interleukin 28B polymorphism and sustained virological response to sofosbuvir plus daclatasvir in chronic hepatitis C genotype 4 Egyptian patients. daclatasvir 103-114 interferon lambda 3 Homo sapiens 20-35 1655654-5 1991 The faster DCVs and the sensory median nerve conduction velocity were significantly slowed in the VWF(+) and VWF(-) groups. daclatasvir 11-15 von Willebrand factor Homo sapiens 98-101 1655654-5 1991 The faster DCVs and the sensory median nerve conduction velocity were significantly slowed in the VWF(+) and VWF(-) groups. daclatasvir 11-15 von Willebrand factor Homo sapiens 109-112 33768560-3 2021 We aimed to evaluate the usefulness of the reference single nucleotide polymorphism (rs12979860) interleukin 28B (CC genotype) for predicting sustained virological response to sofosbuvir plus daclatasvir in Egyptian patients infected with HCV-4. daclatasvir 192-203 interferon lambda 3 Homo sapiens 97-112 33768560-9 2021 WHAT IS NEW AND CONCLUSION: Our results suggest that IL28B genotype contributes to the prediction of response to sofosbuvir plus daclatasvir. daclatasvir 129-140 interferon lambda 3 Homo sapiens 53-58 34890854-1 2022 The separation of daclatasvir and its R,R,R,R-enantiomer was studied by capillary electrophoresis using various randomly methylated beta-CDs and the single isomer heptakis(2,6-di-O-methyl)-beta-CD (2,6-DM-beta-CD) as chiral selectors in an acidic background electrolyte. daclatasvir 18-29 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 189-196 34890854-1 2022 The separation of daclatasvir and its R,R,R,R-enantiomer was studied by capillary electrophoresis using various randomly methylated beta-CDs and the single isomer heptakis(2,6-di-O-methyl)-beta-CD (2,6-DM-beta-CD) as chiral selectors in an acidic background electrolyte. daclatasvir 18-29 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 205-212 34890854-3 2022 HPLC and NMR analyses confirmed that the presence of a high 2,6-DM-beta-CD content in the CDs enables to achieve the migration order R,R,R,R-enantiomer > daclatasvir. daclatasvir 154-165 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 67-74