Skip to main content
Top
Published in: Clinical Pharmacokinetics 9/2013

01-09-2013 | Original Research Article

Pharmacokinetics of a Three-Way Drug Interaction Between Danoprevir, Ritonavir and the Organic Anion Transporting Polypeptide (OATP) Inhibitor Ciclosporin

Authors: Barbara J. Brennan, Sebastian A. Moreira, Peter N. Morcos, Mercidita T. Navarro, Jiney Asthappan, Petra Goelzer, Paul Weigl, Patrick F. Smith

Published in: Clinical Pharmacokinetics | Issue 9/2013

Login to get access

Abstract

Background

Danoprevir (RG7227) is a potent macrocyclic inhibitor of the hepatitis C virus NS3/4A protease, which is currently in development in combination with low-dose ritonavir for the treatment of chronic hepatitis C infection. Danoprevir is a substrate of cytochrome P450 3A4, and the organic anion transporting polypeptides (OATP) 1B1 and 1B3.

Objective

The objective of this study was to evaluate the effect of a potent OATP inhibitor, ciclosporin, on danoprevir pharmacokinetics, when administered as danoprevir/ritonavir. The effect of danoprevir/ritonavir on ciclosporin pharmacokinetics was also investigated.

Methods

This was a single-dose, randomized, open-label, two-sequence, three-period, crossover study in healthy volunteers. In the first period, subjects were randomized to receive either a single oral dose of danoprevir 100 mg in combination with ritonavir 100 mg or a single oral dose of ciclosporin 100 mg. After a 14-day washout, patients were crossed over to receive the opposite treatment. In period 3, all subjects received the combination of danoprevir/ritonavir and ciclosporin following a 14-day washout from period 2. Blood samples were collected serially with each dose for pharmacokinetic assessment. Pharmacokinetic parameters were estimated using non-compartmental analysis. Geometric mean ratios (GMRs) and 90 % confidence intervals (CIs) were used to compare pharmacokinetic parameters [maximum concentration (C max), area under the concentration–time curve from time zero to infinity (AUC), and concentration 12 h post-dose (C 12h)] of danoprevir/ritonavir and ciclosporin when administered alone or in combination. Measures of safety and tolerability were also evaluated.

Results

A total of 18 subjects were enrolled, and 17 completed the study. The C max, AUC, and C 12h GMRs (90 % CI) when danoprevir/ritonavir and ciclosporin were co-administered versus danoprevir/ritonavir or ciclosporin alone were 7.22 (5.42–9.62), 13.6 (11.2–16.6), and 22.5 (17.4–29.3), respectively, for danoprevir, 1.97 (1.72–2.27), 2.23 (2.07–2.42), and 2.50 (2.22–2.81), respectively, for ritonavir, and 1.42 (1.29–1.57), 3.65 (3.27–4.08), and 6.15 (5.32–7.11), respectively, for ciclosporin. All treatments were well tolerated, with no laboratory abnormalities, and no clinically significant changes in vital signs, electrocardiograms, or physical examinations observed.

Conclusions

A significant drug–drug interaction was observed between ciclosporin and danoprevir/ritonavir, leading to substantial increases in exposure to danoprevir and a lesser impact on exposure to ritonavir. Therefore, co-administration of danoprevir/ritonavir with potent OATP inhibitors should be undertaken with appropriate precautions.
Literature
1.
go back to reference Everson GT, Cooper C, Hézode C, et al. High SVR rates with 12 to 24 weeks of ritonavir-boosted danoprevir plus Peg-IFNα-2a (40KD)/RBV in HCV genotype 1 or 4 patients in the DAUPHINE study. Hepatology. 2012;56(4 Suppl):552A. Everson GT, Cooper C, Hézode C, et al. High SVR rates with 12 to 24 weeks of ritonavir-boosted danoprevir plus Peg-IFNα-2a (40KD)/RBV in HCV genotype 1 or 4 patients in the DAUPHINE study. Hepatology. 2012;56(4 Suppl):552A.
2.
go back to reference Gane EJ, Rouzier R, Stedman C, et al. Antiviral activity, safety, and pharmacokinetics of danoprevir/ritonavir plus PEG-IFN α-2a/RBV in hepatitis C patients. J Hepatol. 2011;55(5):972–9.PubMedCrossRef Gane EJ, Rouzier R, Stedman C, et al. Antiviral activity, safety, and pharmacokinetics of danoprevir/ritonavir plus PEG-IFN α-2a/RBV in hepatitis C patients. J Hepatol. 2011;55(5):972–9.PubMedCrossRef
3.
go back to reference Reddy MB, Chen Y, Haznedar JO, et al. Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug–drug interaction study. Clin Pharmacokinet. 2012;51(7):457–65.PubMedCrossRef Reddy MB, Chen Y, Haznedar JO, et al. Impact of low-dose ritonavir on danoprevir pharmacokinetics: results of computer-based simulations and a clinical drug–drug interaction study. Clin Pharmacokinet. 2012;51(7):457–65.PubMedCrossRef
4.
go back to reference Goelzer P, Morcos PN, Tran J, et al. Coadministration of ritonavir with the HCV protease inhibitor danoprevir substantially reduces reactive metabolite formation both in vitro and in vivo. Hepatology. 2012;56(4 Suppl):580A. Goelzer P, Morcos PN, Tran J, et al. Coadministration of ritonavir with the HCV protease inhibitor danoprevir substantially reduces reactive metabolite formation both in vitro and in vivo. Hepatology. 2012;56(4 Suppl):580A.
5.
go back to reference Eley T, Han Y-H, Huang S-P, et al. In vivo and in vitro assessment of asunaprevir as an inhibitor and substrate of OATP transporters in healthy volunteers. Rev Antivir Ther Infect Dis. 2012;6:7. Eley T, Han Y-H, Huang S-P, et al. In vivo and in vitro assessment of asunaprevir as an inhibitor and substrate of OATP transporters in healthy volunteers. Rev Antivir Ther Infect Dis. 2012;6:7.
6.
go back to reference Park S, Ward W, Beaudet B, et al. In vitro assessment of potential drug–drug interactions between telaprevir and cyclophilin inhibitors in the treatment of chronic hepatitis C. Hepatology. 2011;54(Suppl S1):540A. Park S, Ward W, Beaudet B, et al. In vitro assessment of potential drug–drug interactions between telaprevir and cyclophilin inhibitors in the treatment of chronic hepatitis C. Hepatology. 2011;54(Suppl S1):540A.
7.
go back to reference Reddy MB, Morcos PN, Le Pogam S, et al. Pharmacokinetic/pharmacodynamic predictors of clinical potency for hepatitis C virus nonnucleoside polymerase and protease inhibitors. Antimicrob Agents Chemother. 2012;56(6):3144–56.PubMedCrossRef Reddy MB, Morcos PN, Le Pogam S, et al. Pharmacokinetic/pharmacodynamic predictors of clinical potency for hepatitis C virus nonnucleoside polymerase and protease inhibitors. Antimicrob Agents Chemother. 2012;56(6):3144–56.PubMedCrossRef
8.
go back to reference Seiwert SD, Andrews SW, Jiang Y, et al. Preclinical characteristics of the hepatitis C virus NS3/4A protease inhibitor ITMN-191 (R7227). Antimicrob Agents Chemother. 2008;52(12):4432–41.PubMedCrossRef Seiwert SD, Andrews SW, Jiang Y, et al. Preclinical characteristics of the hepatitis C virus NS3/4A protease inhibitor ITMN-191 (R7227). Antimicrob Agents Chemother. 2008;52(12):4432–41.PubMedCrossRef
9.
go back to reference Portmann R, Meille C, Poirier A. Danoprevir: in vitro studies on hepatic active uptake in human hepatocytes. Nutley: Hoffmann-La Roche Inc; 2012 (data on file). Portmann R, Meille C, Poirier A. Danoprevir: in vitro studies on hepatic active uptake in human hepatocytes. Nutley: Hoffmann-La Roche Inc; 2012 (data on file).
10.
go back to reference Wen B, Huang J. In vivo metabolism of danoprevir. Nutley: Hoffmann-La Roche Inc; 2012 (data on file). Wen B, Huang J. In vivo metabolism of danoprevir. Nutley: Hoffmann-La Roche Inc; 2012 (data on file).
11.
go back to reference Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.PubMedCrossRef Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.PubMedCrossRef
12.
go back to reference Shitara Y, Maeda K, Ikejiri K, et al. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78.PubMedCrossRef Shitara Y, Maeda K, Ikejiri K, et al. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78.PubMedCrossRef
13.
go back to reference Saeki T, Ueda K, Tanigawara Y, et al. Human P-glycoprotein transports cyclosporin A and FK506. J Biol Chem. 1993;268:6077–80.PubMed Saeki T, Ueda K, Tanigawara Y, et al. Human P-glycoprotein transports cyclosporin A and FK506. J Biol Chem. 1993;268:6077–80.PubMed
14.
go back to reference Treiber A, Schneiter R, Häusler S, et al. Bosentan is a substrate of human OATP1B1 and OATP1B3: inhibition of hepatic uptake as the common mechanism of its interactions with cyclosporin A, rifampicin, and sildenafil. Drug Metab Dispos. 2007;35(8):1400–7.PubMedCrossRef Treiber A, Schneiter R, Häusler S, et al. Bosentan is a substrate of human OATP1B1 and OATP1B3: inhibition of hepatic uptake as the common mechanism of its interactions with cyclosporin A, rifampicin, and sildenafil. Drug Metab Dispos. 2007;35(8):1400–7.PubMedCrossRef
15.
go back to reference Simonson SG, Raza A, Martin PD, et al. Rosuvastatin pharmacokinetics in heart transplant recipients administered an antirejection regimen including cyclosporine. Clin Pharmacol Ther. 2004;76(2):167–77.PubMedCrossRef Simonson SG, Raza A, Martin PD, et al. Rosuvastatin pharmacokinetics in heart transplant recipients administered an antirejection regimen including cyclosporine. Clin Pharmacol Ther. 2004;76(2):167–77.PubMedCrossRef
16.
go back to reference Park JW, Siekmeier R, Merz M, et al. Pharmacokinetics of pravastatin in heart-transplant patients taking cyclosporin A. Int J Clin Pharmacol Ther. 2002;40(10):439–50.PubMed Park JW, Siekmeier R, Merz M, et al. Pharmacokinetics of pravastatin in heart-transplant patients taking cyclosporin A. Int J Clin Pharmacol Ther. 2002;40(10):439–50.PubMed
17.
go back to reference Binet I, Wallnöfer A, Weber C, et al. Renal hemodynamics and pharmacokinetics of bosentan with and without cyclosporine A. Kidney Int. 2000;57:224–31.PubMedCrossRef Binet I, Wallnöfer A, Weber C, et al. Renal hemodynamics and pharmacokinetics of bosentan with and without cyclosporine A. Kidney Int. 2000;57:224–31.PubMedCrossRef
18.
go back to reference Neoral® (cyclosporine capsules, USP). US prescribing information. New Jersey: Novartis Pharmaceuticals Corporation; 2012. Neoral® (cyclosporine capsules, USP). US prescribing information. New Jersey: Novartis Pharmaceuticals Corporation; 2012.
19.
go back to reference Morcos PN, Moreira S, Navarro M, et al. Ritonavir-boosted danoprevir (DNVr) may be co-administered without regard to meals or in combination with ranitidine or omeprazole: results of a healthy volunteer pharmacokinetic study. Rev Antiviral Ther Infect Dis. 2012;6:7. Morcos PN, Moreira S, Navarro M, et al. Ritonavir-boosted danoprevir (DNVr) may be co-administered without regard to meals or in combination with ranitidine or omeprazole: results of a healthy volunteer pharmacokinetic study. Rev Antiviral Ther Infect Dis. 2012;6:7.
20.
go back to reference Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol. 2009;158(3):693–705.PubMedCrossRef Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol. 2009;158(3):693–705.PubMedCrossRef
21.
go back to reference Ding R, Tayrouz Y, Reidel KD, et al. Substantial pharmacokinetic interaction between digoxin and ritonavir in healthy volunteers. Clin Pharmacol Ther. 2004;76(1):73–84.PubMedCrossRef Ding R, Tayrouz Y, Reidel KD, et al. Substantial pharmacokinetic interaction between digoxin and ritonavir in healthy volunteers. Clin Pharmacol Ther. 2004;76(1):73–84.PubMedCrossRef
22.
go back to reference Vaidyanathan S, Camenisch G, Schuetz H, et al. Pharmacokinetics of the oral direct renin inhibitor aliskiren in combination with digoxin, atorvastatin, and ketoconazole in healthy subjects: the role of P-glycoprotein in the disposition of aliskiren. J Clin Pharmacol. 2008;48(11):1323–38.PubMedCrossRef Vaidyanathan S, Camenisch G, Schuetz H, et al. Pharmacokinetics of the oral direct renin inhibitor aliskiren in combination with digoxin, atorvastatin, and ketoconazole in healthy subjects: the role of P-glycoprotein in the disposition of aliskiren. J Clin Pharmacol. 2008;48(11):1323–38.PubMedCrossRef
23.
go back to reference Keogh J, Kunta J. Development, validation and utility of an in vitro technique for assessment of potential clinical drug–drug interactions involving P-glycoprotein. Eur J Pharm Sci. 2006;27(5):543–54.PubMedCrossRef Keogh J, Kunta J. Development, validation and utility of an in vitro technique for assessment of potential clinical drug–drug interactions involving P-glycoprotein. Eur J Pharm Sci. 2006;27(5):543–54.PubMedCrossRef
24.
go back to reference Reddy MB, Connor A, Brennan BJ, et al. Physiological modeling and assessments of regional drug bioavailability of danoprevir to determine whether a controlled release formulation is feasible. Biopharm Drug Dispos. 2011;32(5):261–75.PubMedCrossRef Reddy MB, Connor A, Brennan BJ, et al. Physiological modeling and assessments of regional drug bioavailability of danoprevir to determine whether a controlled release formulation is feasible. Biopharm Drug Dispos. 2011;32(5):261–75.PubMedCrossRef
25.
go back to reference Asberg A, Hartmann A, Fjeldsa E, et al. Bilateral pharmacokinetic interaction between cyclosporine A and atorvastatin in renal transplant recipients. Am J Transplant. 2001;1(4):382–6.PubMedCrossRef Asberg A, Hartmann A, Fjeldsa E, et al. Bilateral pharmacokinetic interaction between cyclosporine A and atorvastatin in renal transplant recipients. Am J Transplant. 2001;1(4):382–6.PubMedCrossRef
26.
go back to reference Olbricht C, Wanner C, Eisenhauer T, et al. Accumulation of lovastatin, but not pravastatin, in the blood of cyclosporine-treated kidney graft patients after multiple doses. Clin Pharmacol Ther. 1997;62(3):311–21.PubMedCrossRef Olbricht C, Wanner C, Eisenhauer T, et al. Accumulation of lovastatin, but not pravastatin, in the blood of cyclosporine-treated kidney graft patients after multiple doses. Clin Pharmacol Ther. 1997;62(3):311–21.PubMedCrossRef
27.
go back to reference Regazzi MB, Iacona I, Campana C, et al. Altered disposition of pravastatin following concomitant drug therapy with cyclosporin A in transplant recipients. Transplant Proc. 1993;25(4):2732–4.PubMed Regazzi MB, Iacona I, Campana C, et al. Altered disposition of pravastatin following concomitant drug therapy with cyclosporin A in transplant recipients. Transplant Proc. 1993;25(4):2732–4.PubMed
28.
go back to reference Drewe J, Gutmann H, Fricker G, et al. HIV protease inhibitor ritonavir: a more potent inhibitor of P-glycoprotein than the cyclosporine analog SDZ PSC 833. Biochem Pharmacol. 1999;57:1147–52.PubMedCrossRef Drewe J, Gutmann H, Fricker G, et al. HIV protease inhibitor ritonavir: a more potent inhibitor of P-glycoprotein than the cyclosporine analog SDZ PSC 833. Biochem Pharmacol. 1999;57:1147–52.PubMedCrossRef
29.
go back to reference Morcos PN, Chang L, Zhang Y, et al. Danoprevir (DNV) does not change effects of ritonavir (RTV) on the PK of CYP3A substrate midazolam (MDZ) and CYP2C9 substrate warfarin (WAR). Rev Antiviral Ther Infect Dis. 2011;6:4. Morcos PN, Chang L, Zhang Y, et al. Danoprevir (DNV) does not change effects of ritonavir (RTV) on the PK of CYP3A substrate midazolam (MDZ) and CYP2C9 substrate warfarin (WAR). Rev Antiviral Ther Infect Dis. 2011;6:4.
30.
go back to reference Gomez DY, Wacher VJ, Tomlanovich SJ, et al. The effects of ketoconazole on the intestinal metabolism and bioavailability of cyclosporine. Clin Pharmacol Ther. 1995;58(1):15–9.PubMedCrossRef Gomez DY, Wacher VJ, Tomlanovich SJ, et al. The effects of ketoconazole on the intestinal metabolism and bioavailability of cyclosporine. Clin Pharmacol Ther. 1995;58(1):15–9.PubMedCrossRef
31.
go back to reference Hsu A, Granneman GR, Bertz RJ. Ritonavir. Clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet. 1998;35(4):275–91.PubMedCrossRef Hsu A, Granneman GR, Bertz RJ. Ritonavir. Clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet. 1998;35(4):275–91.PubMedCrossRef
32.
go back to reference Vogel M, Voigt E, Michaelis HC, et al. Management of drug-to-drug interactions between cyclosporine A and the protease-inhibitor lopinavir/ritonavir in liver-transplanted HIV-infected patients. Liver Transplant. 2004;10(7):939–44.CrossRef Vogel M, Voigt E, Michaelis HC, et al. Management of drug-to-drug interactions between cyclosporine A and the protease-inhibitor lopinavir/ritonavir in liver-transplanted HIV-infected patients. Liver Transplant. 2004;10(7):939–44.CrossRef
33.
go back to reference Guaraldi G, Cocchi S, Codeluppi M, et al. Pharmacokinetic interaction between amprenavir/ritonavir and FosAmprenavir on cyclosporine in two patients with human immunodeficiency virus infection undergoing orthotopic liver transplantation. Transplant Proc. 2006;38(4):1138–40.PubMedCrossRef Guaraldi G, Cocchi S, Codeluppi M, et al. Pharmacokinetic interaction between amprenavir/ritonavir and FosAmprenavir on cyclosporine in two patients with human immunodeficiency virus infection undergoing orthotopic liver transplantation. Transplant Proc. 2006;38(4):1138–40.PubMedCrossRef
Metadata
Title
Pharmacokinetics of a Three-Way Drug Interaction Between Danoprevir, Ritonavir and the Organic Anion Transporting Polypeptide (OATP) Inhibitor Ciclosporin
Authors
Barbara J. Brennan
Sebastian A. Moreira
Peter N. Morcos
Mercidita T. Navarro
Jiney Asthappan
Petra Goelzer
Paul Weigl
Patrick F. Smith
Publication date
01-09-2013
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 9/2013
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-013-0077-2

Other articles of this Issue 9/2013

Clinical Pharmacokinetics 9/2013 Go to the issue