1
|
Melillo N, Scotcher D, Kenna JG, Green C, Hines CDG, Laitinen I, Hockings PD, Ogungbenro K, Gunwhy ER, Sourbron S, Waterton JC, Schuetz G, Galetin A. Use of In Vivo Imaging and Physiologically-Based Kinetic Modelling to Predict Hepatic Transporter Mediated Drug-Drug Interactions in Rats. Pharmaceutics 2023; 15:896. [PMID: 36986758 PMCID: PMC10057977 DOI: 10.3390/pharmaceutics15030896] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/23/2023] [Accepted: 03/03/2023] [Indexed: 03/12/2023] Open
Abstract
Gadoxetate, a magnetic resonance imaging (MRI) contrast agent, is a substrate of organic-anion-transporting polypeptide 1B1 and multidrug resistance-associated protein 2. Six drugs, with varying degrees of transporter inhibition, were used to assess gadoxetate dynamic contrast enhanced MRI biomarkers for transporter inhibition in rats. Prospective prediction of changes in gadoxetate systemic and liver AUC (AUCR), resulting from transporter modulation, were performed by physiologically-based pharmacokinetic (PBPK) modelling. A tracer-kinetic model was used to estimate rate constants for hepatic uptake (khe), and biliary excretion (kbh). The observed median fold-decreases in gadoxetate liver AUC were 3.8- and 1.5-fold for ciclosporin and rifampicin, respectively. Ketoconazole unexpectedly decreased systemic and liver gadoxetate AUCs; the remaining drugs investigated (asunaprevir, bosentan, and pioglitazone) caused marginal changes. Ciclosporin decreased gadoxetate khe and kbh by 3.78 and 0.09 mL/min/mL, while decreases for rifampicin were 7.20 and 0.07 mL/min/mL, respectively. The relative decrease in khe (e.g., 96% for ciclosporin) was similar to PBPK-predicted inhibition of uptake (97-98%). PBPK modelling correctly predicted changes in gadoxetate systemic AUCR, whereas underprediction of decreases in liver AUCs was evident. The current study illustrates the modelling framework and integration of liver imaging data, PBPK, and tracer-kinetic models for prospective quantification of hepatic transporter-mediated DDI in humans.
Collapse
Affiliation(s)
- Nicola Melillo
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Science, The University of Manchester, Manchester M13 9PL, UK (D.S.)
- SystemsForecastingUK Ltd., Lancaster LA1 5DD, UK
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Science, The University of Manchester, Manchester M13 9PL, UK (D.S.)
| | | | - Claudia Green
- MR & CT Contrast Media Research, Bayer AG, 13353 Berlin, Germany
| | | | - Iina Laitinen
- Sanofi-Aventis Deutschland GmbH, Bioimaging Germany, 65929 Frankfurt am Main, Germany
- Antaros Medical, 431 83 Mölndal, Sweden
| | - Paul D. Hockings
- Antaros Medical, 431 83 Mölndal, Sweden
- MedTech West, Chalmers University of Technology, 413 45 Gothenburg, Sweden
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Science, The University of Manchester, Manchester M13 9PL, UK (D.S.)
| | - Ebony R. Gunwhy
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TA, UK
| | - Steven Sourbron
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TA, UK
| | - John C. Waterton
- Bioxydyn Ltd., Manchester M15 6SZ, UK
- Centre for Imaging Sciences, Division of Informatics Imaging & Data Sciences, School of Health Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Gunnar Schuetz
- MR & CT Contrast Media Research, Bayer AG, 13353 Berlin, Germany
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Science, The University of Manchester, Manchester M13 9PL, UK (D.S.)
| |
Collapse
|
2
|
Al-Nahari MM, Abbassi MM, Ebeid FS, Hassany M, El-Sayed MH, Farid SF. Pharmacokinetics of daclatasvir in Egyptian adolescents with genotype-4 HCV infection. Antivir Ther 2020; 25:101-110. [PMID: 32367815 DOI: 10.3851/imp3357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Daclatasvir has potent antiviral activity against HCV infection when used in combination with sofosbuvir, however, its pharmacokinetics have not been described in adolescents. The aim is to determine the pharmacokinetic parameters of daclatasvir in adolescents, and to develop a population pharmacokinetic (PopPK) model. METHODS Seventeen adolescent patients with genotype-4 chronic HCV infection received once daily oral daclatasvir 60 mg in combination with 400 mg sofosbuvir for 12 weeks. Steady state concentrations were determined. Non-compartmental and population PK were determined. RESULTS The average PK parameters calculated by non-compartmental analysis (NCA): maximum plasma concentration (Cmax), area under the curve (AUC), apparent oral volume of distribution (V/F), apparent oral clearance (CL/F) and half-life (T1/2) were 1,092 ng/ml, 11,178 ng/ml•h, 55 l, 4.5 l/h and 8.5 h, respectively. Daclatasvir was best described by one compartment structural PK model with zero order absorption and first-order elimination. The absorption rate constant (K0), V/F, and CL/F of the final PopPK model of daclatasvir were 1.5/h, 52 l and 4.7 l/h, respectively. Body weight and serum albumin had significant effect on the V/F parameter. CONCLUSIONS Body weight and serum albumin were the major determinants of daclatasvir V/F in this population. PK parameters were comparable to those reported in adult HCV patients, demonstrating that 60 mg daclatasvir is an appropriate dose for adolescents. ClinicalTrials.gov NCT03540212.
Collapse
Affiliation(s)
- Mogeb M Al-Nahari
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Maggie M Abbassi
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Fatma Se Ebeid
- Paediatric Haematology/Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Hassany
- Tropical Medicine Department, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Manal H El-Sayed
- Department of Paediatrics and Clinical Research Centre, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Samar F Farid
- Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Gandhi Y, Eley T, Fura A, Li W, Bertz RJ, Garimella T. Daclatasvir: A Review of Preclinical and Clinical Pharmacokinetics. Clin Pharmacokinet 2019; 57:911-928. [PMID: 29353349 DOI: 10.1007/s40262-017-0624-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Daclatasvir is a first-in-class, highly selective, hepatitis C virus, non-structural protein 5a polymerase replication complex inhibitor with picomolar potency and broad genotypic coverage in vitro. Daclatasvir undergoes rapid absorption, with a time to reach maximum plasma concentration of 1-2 h and an elimination half-life of ~ 10 to 14 h observed in single-ascending dose studies. Steady state was achieved by day 4 in multiple-ascending dose studies. Daclatasvir can be administered without regard to food or pH modifiers. Daclatasvir exposure is similar between healthy subjects and subjects infected with hepatitis C virus. Intrinsic factors such as age, race, or sex do not impact daclatasvir exposure. No dose adjustment is necessary for patients with any degree of hepatic or renal impairment. 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. Renal clearance is a minor route of elimination for daclatasvir. 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. No dose adjustment is required when daclatasvir is co-administered with moderate inhibitors of cytochrome P450 3A4. The dose of daclatasvir should be increased from 60 to 90 mg once daily when co-administered with moderate inducers of cytochrome P450 3A4. Co-administration of daclatasvir with strong inducers of cytochrome P450 3A4 is contraindicated. Concurrent medications with inhibitory effects on P-glycoprotein without concurrent inhibition of cytochrome P450 3A4 are unlikely to cause marked changes in daclatasvir exposure, as the clearance of daclatasvir is through both cytochrome P450 3A4 and P-glycoprotein. The potential for daclatasvir to affect the pharmacokinetics of concomitantly administered drugs that are substrates of the cytochrome P450 enzyme system is low. 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. In summary, daclatasvir is a hepatitis C virus, non-structural protein 5a-selective inhibitor with a well-characterized pharmacokinetic profile that forms part of potent and well-tolerated all-oral treatment regimens for chronic hepatitis C virus infection.
Collapse
Affiliation(s)
- Yash Gandhi
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, NJ, 08540, USA
| | - Timothy Eley
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, NJ, 08540, USA.,Arbutus Biopharma, 701 Veterans Circle, Warminster, PA, 18974, USA
| | - Aberra Fura
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, NJ, 08540, USA
| | - Wenying Li
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, NJ, 08540, USA
| | - Richard J Bertz
- Bristol-Myers Squibb, P.O. Box 4000, Princeton, NJ, 08540, USA
| | | |
Collapse
|
4
|
Safar Z, Kis E, Erdo F, Zolnerciks JK, Krajcsi P. ABCG2/BCRP: variants, transporter interaction profile of substrates and inhibitors. Expert Opin Drug Metab Toxicol 2019; 15:313-328. [PMID: 30856014 DOI: 10.1080/17425255.2019.1591373] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION ABCG2 has a broad substrate specificity and is one of the most important efflux proteins modulating pharmacokinetics of drugs, nutrients and toxicokinetics of toxicants. ABCG2 is an important player in transporter-mediated drug-drug interactions (tDDI). Areas covered: The aims of the review are i) to cover transporter interaction profile of substrates and inhibitors that can be utilized to test interaction of drug candidates with ABCG2, ii) to highlight main characteristics of in vitro testing and iii) to describe the structural basis of the broad substrate specificity of the protein. Preclinical data utilizing Abcg2/Bcrp1 knockouts and clinical studies showing effect of ABCG2 c.421C>A polymorphism on pharmacokinetics of drugs have provided evidence for a broad array of drug substrates and support drug - ABCG2 interaction testing. A consensus on using rosuvastatin and sulfasalazine as intestinal substrates for clinical studies is in the formation. Other substrates relevant to the therapeutic area can be considered. Monolayer efflux assays and vesicular transport assays have been extensively utilized in vitro. Expert opinion: Clinical substrates display complex pharmacokinetics due to broad interaction profiles with multiple transporters and metabolic enzymes. Substrate-dependent inhibition has been observed for several inhibitors. Harmonization of in vitro and in vivo testing makes sense. However, rosuvastatin and sulfasalazine are not efficiently transported in either MDCKII or LLC-PK1-based monolayers. Caco-2 monolayer assays and vesicular transport assays are potential alternatives.
Collapse
Affiliation(s)
| | - Emese Kis
- a SOLVO Biotechnology , Szeged , Hungary
| | - Franciska Erdo
- b Faculty of Information Technology and Bionics , Pázmány Péter Catholic University , Budapest , Hungary
| | | | - Peter Krajcsi
- a SOLVO Biotechnology , Szeged , Hungary.,d Department of Morphology and Physiology. Faculty of Health Sciences , Semmelweis University , Budapest , Hungary
| |
Collapse
|
5
|
Garimella T, Tao X, Sims K, Chang YT, Rana J, Myers E, Wind-Rotolo M, Bhatnagar R, Eley T, LaCreta F, AbuTarif M. 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. Drugs R D 2018; 18:55-65. [PMID: 29255971 PMCID: PMC5833906 DOI: 10.1007/s40268-017-0222-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background A fixed-dose combination of daclatasvir (DCV; hepatitis C virus NS5A inhibitor), asunaprevir (ASV; non-structural protein 3 inhibitor), and beclabuvir (BCV; non-structural protein 5B inhibitor) is approved in Japan for hepatitis C virus genotype 1. Objective The objective of this study was to assess the combination’s drug–drug interaction potential in vivo using a validated cocktail of eight cytochrome P450 (CYP) and transporter probes. Methods We conducted an open-label single-sequence study in healthy adults (n = 20) given single-dose caffeine (CYP1A2 substrate), metoprolol (CYP2D6), flurbiprofen (CYP2C9), montelukast (CYP2C8), omeprazole (CYP2C19), midazolam (CYP3A4), digoxin (P-glycoprotein), and pravastatin (organic anion-transporting polypeptide), alone or with steady-state twice-daily DCV/ASV/BCV 30/200/75 mg (with or without additional BCV 75 mg to adjust for higher exposure in hepatitis C virus infection). Results Daclatasvir/asunaprevir/beclabuvir did not affect CYP1A2, CYP2C8, or CYP2C9; the probe maximum observed concentration and area under the concentration–time curve extrapolated to infinite time geometric mean ratios and 90% confidence intervals were all within the 0.8–1.25 bioequivalence range. Beclabuvir showed moderate dose-dependent CYP2C19 induction; omeprazole maximum observed concentration and area under the concentration–time curve from 0 to the last quantifiable concentration were lower with additional BCV [geometric mean ratio 0.36 (90% confidence interval 0.23–0.55) and 0.34 (0.25–0.46), respectively] than without [0.57 (0.42–0.78), 0.48 (0.39–0.59)]. Weak-to-moderate CYP3A4 induction was observed, plus weak CYP2D6, P-glycoprotein, and organic anion-transporting polypeptide inhibition [maximum observed concentration and area under the concentration–time curve extrapolated to infinite time without additional BCV: midazolam 0.57 (0.50–0.65), 0.53 (0.47–0.60); metoprolol 1.40 (1.20–1.64), 1.71 (1.49–1.97); digoxin 1.23 (1.12–1.35), 1.23 (1.17–1.29); pravastatin 2.01 (1.63–2.47), 1.68 (1.43–1.97)]. Conclusions No dose adjustments with DCV/ASV/BCV are indicated for CYP1A2, CYP2C8, CYP2C9, or P-glycoprotein substrates. CYP3A4, CYP2D6, and OATP substrates should be co-administered with caution. Co-administration with agents solely metabolized by CYP2C19 is not recommended. Electronic supplementary material The online version of this article (10.1007/s40268-017-0222-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tushar Garimella
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA.
| | - Xiaolu Tao
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Karen Sims
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Yi-Ting Chang
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Jignasa Rana
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Elsa Myers
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | | | | | - Timothy Eley
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Frank LaCreta
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Malaz AbuTarif
- Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| |
Collapse
|
6
|
Chan P, Li H, Zhu L, Bifano M, Eley T, Osawa M, Ueno T, Hughes E, Bertz R, Garimella T, AbuTarif M. Population Pharmacokinetic Analysis of Daclatasvir in Subjects with Chronic Hepatitis C Virus Infection. Clin Pharmacokinet 2018; 56:1173-1183. [PMID: 28066880 DOI: 10.1007/s40262-016-0504-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVE Daclatasvir is a potent, pangenotypic once-daily hepatitis C virus (HCV) NS5A inhibitor that is approved for the treatment of chronic HCV infection. The objective of this analysis was to characterize the pharmacokinetics of daclatasvir in subjects with chronic HCV infection. METHODS A population pharmacokinetic (PPK) model was developed to evaluate effects of covariates on daclatasvir pharmacokinetics in subjects with chronic HCV infection (n = 2149 from 11 studies). All significant demographic, laboratory, prognostic and treatment covariates (p < 0.05) from univariate screening were included in the full model. The final model was reached by backward elimination (p < 0.001) and simulations were performed to further evaluate the effects of covariates on daclatasvir exposures. The plasma pharmacokinetics of daclatasvir was described by a two-compartment model with linear elimination. Absorption was modeled as a zero-order release followed by a first-order absorption into the central compartment. RESULTS The typical value of apparent clearance (CL/F) was 5.7 L/h (1.58% relative standard error [RSE]) and of apparent volume of the central compartment (V c/F) was 58.6 L (2.00% RSE). Modest inter-individual variability was estimated for CL/F (35.1%) and V c/F (29.5%). Statistically significant covariates in the final model were sex, race, virus genotype, baseline creatinine clearance, and alanine aminotransferase (ALT) on CL/F and sex, race, and body weight on V c/F. Covariate effects demonstrated a 30% higher area under the plasma concentration-time curve at steady state (AUCss) in female subjects; effects of all other covariates were <16%. CONCLUSIONS The model adequately described the daclatasvir pharmacokinetics and estimated relatively small covariate effects. Considering the exposure range for the therapeutic dose of daclatasvir 60 mg once daily and the favorable safety profile, the small difference in exposures due to these covariates is not considered clinically relevant.
Collapse
Affiliation(s)
- Phyllis Chan
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA.
| | - Hanbin Li
- Quantitative Solutions, Menlo Park, CA, USA
| | - Li Zhu
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Marc Bifano
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Timothy Eley
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | | | | | - Eric Hughes
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Richard Bertz
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Tushar Garimella
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Malaz AbuTarif
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| |
Collapse
|
7
|
The discovery and optimization of naphthalene-linked P2-P4 Macrocycles as inhibitors of HCV NS3 protease. Bioorg Med Chem Lett 2018; 28:43-48. [DOI: 10.1016/j.bmcl.2017.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/29/2017] [Accepted: 11/02/2017] [Indexed: 12/18/2022]
|
8
|
Garimella T, You X, Wang R, Huang SP, Kandoussi H, Bifano M, Bertz R, Eley T. A Review of Daclatasvir Drug-Drug Interactions. Adv Ther 2016; 33:1867-1884. [PMID: 27664109 PMCID: PMC5083780 DOI: 10.1007/s12325-016-0407-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Indexed: 12/28/2022]
Abstract
The treatment of hepatitis C virus (HCV) infection has been revolutionized in recent years by the development of direct-acting antiviral regimens that do not contain peginterferon (pegIFN) and/or ribavirin (RBV). While direct-acting antiviral-based regimens have been shown to be greatly superior to pegIFN/RBV-based regimens in terms of efficacy and safety, they have a greater susceptibility to drug-drug interactions (DDIs). Daclatasvir (DCV)-the benchmark pangenotypic nonstructural protein 5A inhibitor-has been shown to be efficacious and generally well tolerated in partnership with other HCV direct-acting antivirals, including sofosbuvir, asunaprevir (ASV), and ASV plus beclabuvir. 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. This article presents an overview of the drug interaction studies conducted during the clinical development of DCV, the findings of these studies that led to the guidance on concomitant medication use and dosage along with any required DCV dose modifications, and the use of the known metabolic pathway of DCV to guide concomitant dosing where direct drug-drug studies have not been conducted. The robust characterization of the DCV clinical pharmacology program has demonstrated that DCV has few or no clinically relevant DDIs with medications with which it is likely to be co-administered, and the majority of DDIs that do occur can be predicted and easily managed. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
- Tushar Garimella
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA.
| | - Xiaoli You
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Reena Wang
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Shu-Pang Huang
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Hamza Kandoussi
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Marc Bifano
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| | - Richard Bertz
- Bristol-Myers Squibb Research and Development, Hopewell, NJ, USA
| | - Timothy Eley
- Bristol-Myers Squibb Research and Development, Lawrenceville, NJ, USA
| |
Collapse
|
9
|
Desnoyer A, Lê MP, Yazdanpanah Y, Peytavin G. Reply to: "Therapeutic drug monitoring for sofosbuvir and daclatasvir in transplant recipients with chronic hepatitis C and advanced renal disease". J Hepatol 2016; 65:1065-1066. [PMID: 27460834 DOI: 10.1016/j.jhep.2016.07.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 12/04/2022]
Affiliation(s)
- Aude Desnoyer
- Pharmacy Department, Geneva University Hospitals, HUG, Geneva, Switzerland.
| | - Minh Patrick Lê
- Pharmaco-Toxicology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, IAME, INSERM UMR 1137, Paris, France
| | - Yazdan Yazdanpanah
- Infectious Diseases & Tropical Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Gilles Peytavin
- Pharmaco-Toxicology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, IAME, INSERM UMR 1137, Paris, France
| | | |
Collapse
|
10
|
Zhou N, Han Z, Hartman-Neumann S, DeGray B, Ueland J, Vellucci V, Hernandez D, McPhee F. Characterization of NS5A polymorphisms and their impact on response rates in patients with HCV genotype 2 treated with daclatasvir-based regimens. J Antimicrob Chemother 2016; 71:3495-3505. [PMID: 27605597 DOI: 10.1093/jac/dkw336] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/22/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Daclatasvir (DCV) is a pan-genotypic non-structural protein 5A (NS5A) inhibitor that is approved for treatment of hepatitis C virus (HCV) genotype (GT)1 and GT3 in the USA and GT1, GT3 and GT4 in Europe. We set out to examine the impact of daclatasvir-based regimens on the sustained virologic response (SVR) in patients with GT2 infection with respect to GT2 subtype and NS5A polymorphisms at amino acid positions associated with daclatasvir resistance. METHODS Analyses were performed on 283 GT2 NS5A sequences from five daclatasvir regimen-based clinical trials (ClinicalTrials.gov: NCT-01257204, NCT-01359644, NCT-02032875, NCT-02032888 and NCT-01616524) and 143 NS5A sequences from the Los Alamos HCV database. Susceptibility analyses of substitutions at amino acid positions associated with daclatasvir resistance and patient-derived NS5A sequences were performed using an in vitro HCV replication assay. RESULTS Of 13 GT2 subtypes identified from 426 NS5A sequences, the most prevalent were GT2a (32%), GT2b (48%) and GT2c (10%). The most prevalent NS5A polymorphism was L31M (GT2a = 88%; GT2b = 59%; GT2c = 10%). Substitutions identified in 96% of GT2 NS5A sequences exhibited daclatasvir EC50 values ranging from 0.005 to 20 nM when tested in vitro. A similar range in daclatasvir EC50 values was observed for 16 diverse GT2 patient-derived NS5A sequences (EC50 = 0.005-60 nM). Depending on the daclatasvir-based regimen studied (daclatasvir/interferon-based or daclatasvir/sofosbuvir-based), SVR rates ranged from 90% to 100% in GT2 patients with the most prevalent baseline NS5A-L31M polymorphism, compared with from 96% to 100% without this polymorphism. CONCLUSIONS High SVR rates were achieved in patients infected with GT2 treated with daclatasvir-based regimens irrespective of GT2 subtype or baseline NS5A polymorphisms.
Collapse
Affiliation(s)
- Nannan Zhou
- Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| | - Zhou Han
- Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| | | | - Brenda DeGray
- Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| | - Joseph Ueland
- Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| | | | | | - Fiona McPhee
- Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| |
Collapse
|
11
|
Desnoyer A, Pospai D, Lê MP, Gervais A, Heurgué-Berlot A, Laradi A, Harent S, Pinto A, Salmon D, Hillaire S, Fontaine H, Zucman D, Simonpoli AM, Muret P, Larrouy L, Bernard Chabert B, Descamps D, Yazdanpanah Y, Peytavin G. Pharmacokinetics, safety and efficacy of a full dose sofosbuvir-based regimen given daily in hemodialysis patients with chronic hepatitis C. J Hepatol 2016; 65:40-47. [PMID: 26952005 DOI: 10.1016/j.jhep.2016.02.044] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV) infection is an independent risk factor for chronic kidney disease and leads to faster liver disease progression in patients requiring hemodialysis than in those with normal renal function. Little is known about the use of a sofosbuvir-containing regimen for infected patients on hemodialysis. We aimed to describe the pharmacokinetics, safety and efficacy of sofosbuvir in 2 dosing regimens and associated antiviral agents in HCV-infected patients requiring hemodialysis. METHODS Multicenter, prospective and observational study of patients receiving sofosbuvir, 400mg once daily (n=7) or 3 times a week (n=5), after hemodialysis with simeprevir, daclatasvir, ledipasvir or ribavirin was conducted. Drug plasma concentrations were determined by liquid chromatography-tandem mass spectrometry before and after a 4h hemodialysis and 1.5h after last drug intake at the end of hemodialysis. RESULTS Plasma concentrations of sofosbuvir or its inactive metabolite sofosbuvir-007 did not accumulate with either regimen between hemodialysis sessions or throughout the treatment course. Sofosbuvir-007 extraction ratio (52%) was consistent with historical data. In one patient receiving the once daily regimen, sofosbuvir-007 half-life was slightly higher (38h) than for patients with normal renal function receiving a full dose. Hemodialysis did not remove any other associated anti-HCV agents. Clinical and biological tolerance was good for all patients. Two relapses occurred with the 3 times a week regimen and none with the once daily. CONCLUSIONS A regimen including sofosbuvir, 400mg once daily, could be proposed for HCV-infected patients requiring hemodialysis and should be associated with close clinical, biological, cardiovascular, and therapeutic drug monitoring. LAY SUMMARY Hepatitis C Virus (HCV) infection in hemodialysis patients is prevalent and aggressive. Effective anti-HCV treatment in these patients may stabilize their renal disease. However, sofosbuvir, the cornerstone of most anti-HCV-containing regimens, should not be administered to these patients until more data is available. In this pharmacokinetic study, sofosbuvir full dose (400mg once daily) administered every day with another direct antiviral agent did not accumulate in hemodialysis patients and was safe and effective.
Collapse
Affiliation(s)
- Aude Desnoyer
- Pharmaco-Toxicology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France.
| | - Dan Pospai
- Gastroenterology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Minh Patrick Lê
- Pharmaco-Toxicology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Anne Gervais
- Infectious Diseases & Tropical Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | | | | | - Stanislas Harent
- Infectious Diseases & Tropical Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Adriana Pinto
- Infectious Diseases & Tropical Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Dominique Salmon
- Internal Medicine Department, Cochin Hospital, APHP, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Sophie Hillaire
- Gastroenterology Department, Foch Hospital, Suresnes, France
| | - Hélène Fontaine
- Gastroenterology Department, Cochin Hospital, APHP, Paris Descartes University, Sorbonne Paris Cité, Inserm U1016, Paris, France
| | - David Zucman
- Infectious Diseases Department, Foch Hospital, Suresnes, France
| | | | - Patrice Muret
- Clinical Pharmaco-Toxicology Department, University Hospital of Besançon, INSERM U1098, Besançon, France
| | - Lucile Larrouy
- Virology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, IAME, INSERM UMR 1137, Paris, France
| | | | - Diane Descamps
- Virology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, IAME, INSERM UMR 1137, Paris, France
| | - Yazdan Yazdanpanah
- Infectious Diseases & Tropical Department, Bichat-Claude Bernard Hospital, APHP, Paris, France
| | - Gilles Peytavin
- Pharmaco-Toxicology Department, Bichat-Claude Bernard Hospital, APHP, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, IAME, INSERM UMR 1137, Paris, France
| |
Collapse
|
12
|
Smith MA, Regal RE, Mohammad RA. Daclatasvir: A NS5A Replication Complex Inhibitor for Hepatitis C Infection. Ann Pharmacother 2015; 50:39-46. [PMID: 26486762 DOI: 10.1177/1060028015610342] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES To review the pharmacology, efficacy, and safety of daclatasvir in the treatment of patients with chronic hepatitis C virus (HCV) infection. DATA SOURCES A literature search through EMBASE and PubMed was conducted (January 1966 to August 2015) using the terms BMS-790052, daclatasvir, and hepatitis C. References from retrieved articles were reviewed for any additional material. Additionally, the new drug application and prescribing information were retrieved. STUDY SELECTION/DATA EXTRACTION The literature search was limited to human studies published in English. Phase 1, 2, and 3 studies describing the pharmacology, pharmacokinetics, efficacy, and safety of daclatasvir for HCV were identified. DATA SYNTHESIS Daclatasvir, a nonstructural 5A protein inhibitor, combined with sofosbuvir, is indicated for adult patients with chronic HCV genotype 3 regardless of treatment or cirrhosis status. The phase III ALLY-3 trial (n = 152) demonstrated that daclatasvir taken once daily with sofosbuvir for 12 weeks was effective at achieving sustained virological response (SVR) rates in treatment-naïve (97%) and treatment-experienced (94%) patients without cirrhosis. Patients with cirrhosis had significantly lower SVR rates (58 and 69%, respectively). The most common adverse drug events associated with daclatasvir and sofosbuvir in ALLY-3 were headache (20%), fatigue (19%), and nausea (12%). CONCLUSIONS Daclatasvir, when combined with sofosbuvir, is an effective agent to treat HCV genotype 3, with SVR rates above 90% for patients without cirrhosis who are treatment naïve or experienced. SVR rates for treatment-naïve or -experienced patients with cirrhosis are not as robust (58%-69%).
Collapse
|
13
|
Raltegravir Pharmacokinetics in Patients on Asunaprevir-Daclatasvir. Antimicrob Agents Chemother 2015; 59:7903-5. [PMID: 26438504 DOI: 10.1128/aac.01603-15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/01/2015] [Indexed: 12/28/2022] Open
Abstract
Raltegravir pharmacokinetics was studied in 20 patients included in the ANRS HC30 QUADRIH Study before and after addition of anti-hepatitis C virus (anti-HCV) quadritherapy, including pegylated-interferon-ribavirin and asunaprevir plus daclatasvir. Raltegravir pharmacokinetic parameters remained unchanged whether administered on or off anti-HCV therapy. In addition, concentrations of raltegravir, asunaprevir, and daclatasvir were not affected by liver cirrhosis. These data suggest that in human immunodeficiency virus (HIV)-HCV-coinfected patients, whether cirrhotic or not, asunaprevir and daclatasvir could be administered safely with raltegravir.
Collapse
|
14
|
Akamatsu N, Sugawara Y, Kokudo N. Asunaprevir (BMS-650032) for the treatment of hepatitis C virus. Expert Rev Anti Infect Ther 2015; 13:1307-17. [PMID: 26414905 DOI: 10.1586/14787210.2015.1091724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Asunaprevir, a second-generation NS3 protease inhibitor of hepatitis C virus (HCV), exhibits strong antiviral activity against HCV genotypes 1 and 4, but relatively weak activity against genotypes 2 and 3. For chronic HCV infection, asunaprevir with daclatasvir as an interferon-free dual treatment achieves a sustained virologic response of nearly 90% in genotype 1b, and a triple regimen with beclabuvir achieves an sustained virologic response >90%. Asunaprevir and daclatasvir dual treatment can be safely and effectively administered to liver transplant recipients with recurrent HCV. The major drawback of asunaprevir is its low threshold to resistance, which can be overcome by combining it with other direct-acting antivirals. Further studies of asunaprevir in combination with other direct-acting antivirals for the treatment of patients with HCV genotypes 1 or 4 and renal impairment or end-stage renal disease under hemodialysis, HIV-coinfection and liver and/or kidney transplant recipients are warranted.
Collapse
Affiliation(s)
- Nobuhisa Akamatsu
- a 1 Artificial Organ and Transplantation Surgery Division, Department of Surgery, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yasuhiko Sugawara
- b 2 Divisions of Hepato-Biliary-Pancreatic Surgery and Liver transplantation, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo, 150-8935, Japan
| | - Norihiro Kokudo
- a 1 Artificial Organ and Transplantation Surgery Division, Department of Surgery, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
15
|
Gentile I, Zappulo E, Buonomo AR, Scotto R, Borgia G. Asunaprevir for hepatitis C: a safety evaluation. Expert Opin Drug Saf 2015; 14:1631-46. [PMID: 26329454 DOI: 10.1517/14740338.2015.1084287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The introduction of direct-acting antiviral (DAA) agents has revolutionized the treatment of hepatitis C virus (HCV) chronic infection. Non-structural 3 protease inhibitors are currently the most numerous class of DAAs on the market. AREAS COVERED This review mainly focuses on the tolerability and safety profile of asunaprevir (ASV)-containing DAA regimens. ASV is a second-wave protease inhibitor currently in Phase III clinical development in most countries and already available in Japan. EXPERT OPINION ASV shows potent antiviral effect and clinical efficacy on HCV genotypes 1 and 4. The all-oral combination daclatasvir/ASV reached high eradication rates in HCV genotype 1b and 4 infection, and a lower efficacy in genotype 1a infection. ASV presents a low potential for drug-drug interaction and a good tolerability as part of multiple, including all-oral, regimens. ASV is associated with a transient and usually mild increase in aminotransferase levels in a low percentage of cases. Due to the impaired pharmacokinetic profile observed in advanced liver disease, ASV use in patients with moderate or severe hepatic impairment is not allowed. In conclusion, ASV represents a powerful weapon against HCV infection and has to be considered an optimal option as a component of genotype tailored interferon-free combinations.
Collapse
Affiliation(s)
- Ivan Gentile
- a University of Naples "Federico II", Department of Clinical Medicine and Surgery , via S. Pansini 5, I-80131 Naples, Italy +39 081 746 3178 ; +39 081 746 3190 ;
| | - Emanuela Zappulo
- a University of Naples "Federico II", Department of Clinical Medicine and Surgery , via S. Pansini 5, I-80131 Naples, Italy +39 081 746 3178 ; +39 081 746 3190 ;
| | - Antonio Riccardo Buonomo
- a University of Naples "Federico II", Department of Clinical Medicine and Surgery , via S. Pansini 5, I-80131 Naples, Italy +39 081 746 3178 ; +39 081 746 3190 ;
| | - Riccardo Scotto
- a University of Naples "Federico II", Department of Clinical Medicine and Surgery , via S. Pansini 5, I-80131 Naples, Italy +39 081 746 3178 ; +39 081 746 3190 ;
| | - Guglielmo Borgia
- a University of Naples "Federico II", Department of Clinical Medicine and Surgery , via S. Pansini 5, I-80131 Naples, Italy +39 081 746 3178 ; +39 081 746 3190 ;
| |
Collapse
|
16
|
Mosure KW, Knipe JO, Browning M, Arora V, Shu YZ, Phillip T, Mcphee F, Scola P, Balakrishnan A, Soars MG, Santone K, Sinz M. Preclinical Pharmacokinetics and In Vitro Metabolism of Asunaprevir (BMS-650032), a Potent Hepatitis C Virus NS3 Protease Inhibitor. J Pharm Sci 2015; 104:2813-23. [DOI: 10.1002/jps.24356] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/23/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022]
|
17
|
Asunaprevir: A Review of Preclinical and Clinical Pharmacokinetics and Drug–Drug Interactions. Clin Pharmacokinet 2015; 54:1205-22. [DOI: 10.1007/s40262-015-0299-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
18
|
Furihata T, Fu Z, Suzuki Y, Matsumoto S, Morio H, Tsubota A, Matsumoto S, Chiba K. Differential inhibition features of direct-acting anti-hepatitis C virus agents against human organic anion transporting polypeptide 2B1. Int J Antimicrob Agents 2015; 46:381-8. [PMID: 26163159 DOI: 10.1016/j.ijantimicag.2015.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/19/2015] [Accepted: 05/05/2015] [Indexed: 11/29/2022]
Abstract
Simeprevir (SMV), asunaprevir (ASV), daclatasvir (DCV) and sofosbuvir (SOF), which are direct-acting antiviral (DAA) agents, are expected to become essential pharmaceutical tools in the fight against the hepatitis C virus (HCV). However, because DAAs are taken orally, there is a potential risk of drug-drug interactions (DDIs) at the absorption step with co-administered drugs in the small intestine. Since it is known that organic anion transporting polypeptide 2B1 (OATP2B1) is one of the key transporters contributing to intestinal drug absorption, it is important to thoroughly understand the inhibition profiles of various DAAs in relation to OATP2B1 function in order to avoid unexpected DDIs. Therefore, using a cell-based transport assay, this study aimed at clarifying such DAA inhibition characteristics towards OATP2B1 function. The results of co-incubation inhibition assays showed that SMV and ASV strongly inhibited estrone sulfate (5 nM) uptake by OATP2B1, with half maximal inhibitory concentrations of 0.49 ± 0.12 μM and 0.16 ± 0.06 μM, respectively. Furthermore, it was found that SMV and ASV imposed long-lasting pre-incubation inhibitory effects on OATP2B1 function that enhanced their co-incubation inhibition potencies. On the other hand, no (or much less significant) inhibitory effects were observed for SOF or DCV. To summarise, these results show that SMV and ASV are co-incubation, as well as long-lasting pre-incubation, inhibitors of OATP2B1 function and therefore these inhibitions may lead to clinically relevant DDIs when used with OATP2B1 substrates.
Collapse
Affiliation(s)
- Tomomi Furihata
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan.
| | - Zhongguo Fu
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Yuki Suzuki
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Shogo Matsumoto
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Hanae Morio
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Akihito Tsubota
- Core Research Facilities for Basic Science (Division of Molecular Cell Biology), Research Center for Medical Science, The Jikei University School of Medicine, 3-19-18 Nishi-Shinbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Sayaka Matsumoto
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Kan Chiba
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| |
Collapse
|