Skip to main content
Log in

Elucidating the Plasma and Liver Pharmacokinetics of Simeprevir in Special Populations Using Physiologically Based Pharmacokinetic Modelling

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

The disposition of simeprevir (SMV) in humans is characterised by cytochrome P450 3A4 metabolism and hepatic uptake by organic anion transporting polypeptide 1B1/3 (OATP1B1/3). This study was designed to investigate SMV plasma and liver exposure upon oral administration in subjects infected with hepatitis C virus (HCV), in subjects of Japanese or Chinese origin, subjects with organ impairment and subjects with OATP genetic polymorphisms, using physiologically based pharmacokinetic modelling. Simulations showed that compared with healthy Caucasian subjects, SMV plasma exposure was 2.4-, 1.7-, 2.2- and 2.0-fold higher, respectively, in HCV-infected Caucasian subjects, in healthy Japanese, healthy Chinese and subjects with severe renal impairment. Further simulations showed that compared with HCV-infected Caucasian subjects, SMV plasma exposure was 1.6-fold higher in HCV-infected Japanese subjects. In subjects with OATP1B1 genetic polymorphisms, no noteworthy changes in SMV pharmacokinetics were observed. Simulations suggested that liver concentrations in Caucasians with HCV are 18 times higher than plasma concentrations.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Janssen Research and Development. Olysio™ (SMV): US prescribing information. http://www.olysio.com/shared/product/olysio/prescribing-information.pdf. Accessed 27 May 2015.

  2. Ouwerkerk-Mahadevan S, Sekar V, Simion A, et al. The pharmacokinetic interactions of the HCV protease inhibitor SMV (TMC435) with HIV antiretroviral agents in healthy volunteers. Poster 1618 presented at the ID week; 17–21 Oct 2012; San Diego (CA).

  3. Ouwerkerk-Mahadevan S, Snoeys J, Peeters M, et al. Drug–drug interactions with the NS3/4A protease inhibitor simeprevir. Clin Pharmacokinet. 2016;55(2):197–208.

    Article  CAS  PubMed  Google Scholar 

  4. Ouwerkerk-Mahadevan S, Snoeys J, Simion A, et al. A novel microdose approach to assess bioavailability, intestinal absorption, gut metabolism, and hepatic clearance of SMV in healthy volunteers. Poster oral presentation presented at the 15th international workshop on clinical pharmacology of HIV and hepatitis therapy, 19–21 May 2014, Washington, DC.

  5. Reesink HW, Fanning GC, Farha KA, et al. Rapid HCV-RNA decline with once daily TMC435: a phase I study in healthy volunteers and hepatitis C patients. Gastroenterology. 2010;138:913–21.

    Article  CAS  PubMed  Google Scholar 

  6. Snoeys J, Beumont M, Monshouwer M, Ouwerkerk-Mahadevan S. A mechanistic understanding of the non-linear pharmacokinetics and intersubject variability of SMV: a PBPK-guided drug development approach. Clin Pharmacol Ther. 2016;99:224–34.

    Article  CAS  PubMed  Google Scholar 

  7. Barter ZE, Tucker GT, Rowland-Yeo K. Differences in cytochrome p450-mediated pharmacokinetics between Chinese and Caucasian populations predicted by mechanistic physiologically based pharmacokinetic modelling. Clin Pharmacokinet. 2013;52(12):1085–100.

    Article  CAS  PubMed  Google Scholar 

  8. Inoue S, Howgate EM, Rowland-Yeo K, et al. Prediction of in vivo drug clearance from in vitro data. II: potential inter-ethnic differences. Xenobiotica. 2006;36(6):499–513.

    Article  CAS  PubMed  Google Scholar 

  9. Rowland Yeo K, Aarabi M, Jamei M, Rostami-Hodjegan A. Modeling and predicting drug pharmacokinetics in patients with renal impairment. Expert Rev Clin Pharmacol. 2011;4(2):261–74.

    Article  PubMed  Google Scholar 

  10. Johnson TN, Boussery K, Rowland-Yeo K, et al. A semi-mechanistic model to predict the effects of liver cirrhosis on drug clearance. Clin Pharmacokinet. 2010;49(3):189–206.

    Article  CAS  PubMed  Google Scholar 

  11. Lin, et al. Liver volume in patients with or without chronic liver diseases. Hepatogastroenterology. 1998;45:1069–74.

    CAS  PubMed  Google Scholar 

  12. Nakai K, Tanaka H, Hanada K, et al. Decreased expression of cytochromes P450 1A2, 2E1, 3A4 and drug transporters Na+-taurocholate-cotransporting polypeptide, organic cation transporter 1, and organic anion-transporting peptide-C correlates with the progression of liver fibrosis in chronic hepatitis C patients. Drug Metab Dispos. 2008;36:1786–93.

    Article  CAS  PubMed  Google Scholar 

  13. Ohnishi A, Murakami S, Akizuki S, et al. In vivo metabolic activity of CYP2C19 and CYP3A in relation to CYP2C19 genetic polymorphism in chronic liver disease. J Clin Pharmacol. 2005;5:1221–9.

    Article  Google Scholar 

  14. Barreiro P, Rodriguez-Novoa S, Labarga P, et al. Influence of liver fibrosis stage on plasma levels of antiretroviral drugs in HIV-infected patients with chronic hepatitis C. J Infect Dis. 2007;195:973–9.

    Article  Google Scholar 

  15. Knox, et al. Ritonavir greatly impairs CYP3A activity in HIV infection with chronic viral hepatitis. J Acquir Immune Defic Syndr. 2008;49:358–68.

    Article  CAS  PubMed  Google Scholar 

  16. Abdallah, et al. Effect of hepatic impairment on the multiple-dose pharmacokinetics of ranolazine sustained-release tablets. J Clin Pharmacol. 2005;45:802–9.

    Article  CAS  PubMed  Google Scholar 

  17. Tomita Y, Maeda K, Sugiyama Y. Ethnic variability in the plasma exposures of OATP1B1 substrates such as HMG-CoA reductase inhibitors: a kinetic consideration of its mechanism. Clin Pharmacol Ther. 2013;94(1):37–51.

    Article  CAS  PubMed  Google Scholar 

  18. Shitara Y, Maeda K, Ikejiri K, et al. Clinical significance of organic anion transporting polypeptides (OATPs) in drug disposition: their roles in the hepatic clearance and intestinal absorption. Biopharm Drug Dispos. 2013;34(1):45–78.

    Article  CAS  PubMed  Google Scholar 

  19. Ouwerkerk-Mahadevan S, Beumont-Mauviel M, Mortier S, et al. Evaluation of the pharmacokinetics and renal excretion of simeprevir in subjects with renal impairment. Drugs R&D. 2015;15:261–70.

    Article  CAS  Google Scholar 

  20. Ouwerkerk-Mahadevan S, Simion A, Spittaels K, Beumont-Mauviel M. Pharmacokinetics of simeprevir (TMC435) in volunteers with moderate or severe hepatic impairment. Oral presentation at the EASL 48th annual meeting, 24–28 April 2013, Amsterdam.

  21. Morcos PN, Chang L, Kulkarni R, et al. A randomised study of the effect of danoprevir/ritonavir or ritonavir on substrates of cytochrome P450 (CYP) 3A and 2C9 in chronic hepatitis C patients using a drug cocktail. Eur J Clin Pharmacol. 2013;69(11):1939–49.

    Article  CAS  PubMed  Google Scholar 

  22. Mathias AA, German P, Murray BP, et al. Pharmacokinetics and pharmacodynamics of GS-9350: a novel pharmacokinetic enhancer without anti-HIV activity. Clin Pharmacol Ther. 2010;87(3):322–9.

    Article  CAS  PubMed  Google Scholar 

  23. Wei L, Han T, Yang D, et al. TIGER team. Simeprevir plus peginterferon/ribavirin for HCV genotype 1-infected treatment-naïve patients in China and South Korea. J Gastroenterol Hepatol. 2016;31(5):912–20.

    Article  CAS  PubMed  Google Scholar 

  24. Niemi M, Schaeffeler E, Lang T, et al. High plasma pravastatin concentrations are associated with single nucleotide polymorphisms and haplotypes of organic anion transporting polypeptide-C (OATP-C, SLCO1B1). Pharmacogenetics. 2004;14:429–40.

    Article  CAS  PubMed  Google Scholar 

  25. Yoshida K, Sun B, Zhang L, et al. Systematic and quantitative assessment of the effect of chronic kidney disease on CYP2D6 and CYP3A4/5. Clin Pharmacol Ther. 2016;100(1):75–87.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Center for Drug Evaluation and Research, US FDA. Clinical pharmacology and biopharmaceutics review(s). http://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/205123Orig1s000ClinPharmR.pdf. Accessed 29 Oct 2016.

  27. European Medicines Agency. Assessment report: Olysio. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_Public_assessment_report/human/002777/WC500167870.pdf. Accessed 29 Oct 2016.

Download references

Acknowledgements

Medical writing assistance was provided by Manoranjenni Chetty of Certara and editorial support was provided by Kimberley Haines of Complete Medical Communications, both funded by Janssen.

Author contributions

MB, MM and S O-M provided scientific input in the clinical studies, analysis of the data and contributed to the writing of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jan Snoeys.

Ethics declarations

Conflict of interest

JS, MB, MM and S O-M are employees of Janssen and may own stock options in the company.

Funding

No funding was received for the preparation of the article.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Snoeys, J., Beumont, M., Monshouwer, M. et al. Elucidating the Plasma and Liver Pharmacokinetics of Simeprevir in Special Populations Using Physiologically Based Pharmacokinetic Modelling. Clin Pharmacokinet 56, 781–792 (2017). https://doi.org/10.1007/s40262-016-0476-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-016-0476-2

Keywords

Navigation