Skip to main content
Top
Published in: Clinical Pharmacokinetics 10/2018

01-10-2018 | Original Research Article

Predictive Performance of Physiologically-Based Pharmacokinetic Models in Predicting Drug–Drug Interactions Involving Enzyme Modulation

Authors: Chia-Hsiang Hsueh, Vicky Hsu, Yuzhuo Pan, Ping Zhao

Published in: Clinical Pharmacokinetics | Issue 10/2018

Login to get access

Abstract

Background

Physiologically-based pharmacokinetic (PBPK) modeling in predicting metabolic drug–drug interactions (mDDIs) is routinely used in drug development. Currently, the US FDA endorses the use of PBPK to potentially support dosing recommendations for investigational drugs as enzyme substrates of mDDIs, and to inform a lack of mDDIs for investigational drugs as enzyme modulators.

Methods

We systematically evaluated the performance of PBPK modeling in predicting mDDIs published in the literature. Models developed to assess both investigational drugs as enzyme substrates (Groups 1 and 2, as being inhibited and induced, respectively) or enzyme modulators (Groups 3 and 4, as inhibitors and inducers, respectively) were evaluated. Predicted ratios of the area under the curve (AUCRs) and/or maximum plasma concentration (CmaxRs) with and without comedication were compared with the observed ratios.

Results

For Groups 1, 2, 3, and 4, 62, 50, 44, and 43% of model-predicted AUCRs, respectively, were within a predefined threshold of 1.25-fold of observed values (0.8–1.25x). When the threshold was widened to twofold, the values increased to 100, 80, 81, and 86% (0.5–2.0x). For Groups 3 and 4, prediction for mDDI liability (the existence or lack of mDDIs) using PBPK appears to be satisfactory.

Conclusion

Our analysis supports the FDA’s current recommendations on the use of PBPK to predict mDDIs.
Appendix
Available only for authorised users
Literature
3.
go back to reference US Food and Drug Administration. US Food and Drug Administration Pharmaceutical Science and Clinical Pharmacology Advisory Committee Meeting. Silver Spring: US FDA; 2017. US Food and Drug Administration. US Food and Drug Administration Pharmaceutical Science and Clinical Pharmacology Advisory Committee Meeting. Silver Spring: US FDA; 2017.
4.
go back to reference Sager JE, Yu J, Ragueneau-Majlessi I, Isoherranen N. Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches: a systematic review of published models, applications, and model verification. Drug Metab Dispos. 2015;43(11):1823–37.CrossRefPubMedPubMedCentral Sager JE, Yu J, Ragueneau-Majlessi I, Isoherranen N. Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches: a systematic review of published models, applications, and model verification. Drug Metab Dispos. 2015;43(11):1823–37.CrossRefPubMedPubMedCentral
5.
go back to reference Wagner C, Zhao P, Pan Y, Hsu V, Grillo J, Huang SM, et al. Application of physiologically based pharmacokinetic (PBPK) modeling to support dose selection: report of an FDA public workshop on PBPK. CPT Pharmacometr Syst Pharmacol. 2015;4(4):226–30.CrossRef Wagner C, Zhao P, Pan Y, Hsu V, Grillo J, Huang SM, et al. Application of physiologically based pharmacokinetic (PBPK) modeling to support dose selection: report of an FDA public workshop on PBPK. CPT Pharmacometr Syst Pharmacol. 2015;4(4):226–30.CrossRef
6.
go back to reference Wagner C, Pan Y, Hsu V, Grillo JA, Zhang L, Reynolds KS, et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin Pharmacokinet. 2015;54(1):117–27.CrossRefPubMed Wagner C, Pan Y, Hsu V, Grillo JA, Zhang L, Reynolds KS, et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin Pharmacokinet. 2015;54(1):117–27.CrossRefPubMed
7.
go back to reference Wagner C, Pan Y, Hsu V, Sinha V, Zhao P. Predicting the effect of CYP3A inducers on the pharmacokinetics of substrate drugs using physiologically based pharmacokinetic (PBPK) modeling: an analysis of PBPK submissions to the US FDA. Clin Pharmacokinet. 2016;55(4):475–83.CrossRefPubMed Wagner C, Pan Y, Hsu V, Sinha V, Zhao P. Predicting the effect of CYP3A inducers on the pharmacokinetics of substrate drugs using physiologically based pharmacokinetic (PBPK) modeling: an analysis of PBPK submissions to the US FDA. Clin Pharmacokinet. 2016;55(4):475–83.CrossRefPubMed
9.
go back to reference Einolf HJ, Chen L, Fahmi OA, Gibson CR, Obach RS, Shebley M, et al. Evaluation of various static and dynamic modeling methods to predict clinical CYP3A induction using in vitro CYP3A4 mRNA induction data. Clin Pharmacol Ther. 2014;95(2):179–88.CrossRefPubMed Einolf HJ, Chen L, Fahmi OA, Gibson CR, Obach RS, Shebley M, et al. Evaluation of various static and dynamic modeling methods to predict clinical CYP3A induction using in vitro CYP3A4 mRNA induction data. Clin Pharmacol Ther. 2014;95(2):179–88.CrossRefPubMed
10.
go back to reference Fahmi OA, Shebley M, Palamanda J, Sinz MW, Ramsden D, Einolf HJ, et al. Evaluation of CYP2B6 induction and prediction of clinical drug–drug interactions: considerations from the IQ consortium induction working group—an industry perspective. Drug Metab Dispos. 2016;44(10):1720–30.CrossRefPubMed Fahmi OA, Shebley M, Palamanda J, Sinz MW, Ramsden D, Einolf HJ, et al. Evaluation of CYP2B6 induction and prediction of clinical drug–drug interactions: considerations from the IQ consortium induction working group—an industry perspective. Drug Metab Dispos. 2016;44(10):1720–30.CrossRefPubMed
11.
go back to reference Guest EJ, Aarons L, Houston JB, Rostami-Hodjegan A, Galetin A. Critique of the two-fold measure of prediction success for ratios: application for the assessment of drug–drug interactions. Drug Metab Dispos. 2011;39(2):170–3.CrossRefPubMed Guest EJ, Aarons L, Houston JB, Rostami-Hodjegan A, Galetin A. Critique of the two-fold measure of prediction success for ratios: application for the assessment of drug–drug interactions. Drug Metab Dispos. 2011;39(2):170–3.CrossRefPubMed
12.
go back to reference Vieira MD, Kim MJ, Apparaju S, Sinha V, Zineh I, Huang SM, et al. PBPK model describes the effects of comedication and genetic polymorphism on systemic exposure of drugs that undergo multiple clearance pathways. Clin Pharmacol Ther. 2014;95(5):550–7.CrossRefPubMed Vieira MD, Kim MJ, Apparaju S, Sinha V, Zineh I, Huang SM, et al. PBPK model describes the effects of comedication and genetic polymorphism on systemic exposure of drugs that undergo multiple clearance pathways. Clin Pharmacol Ther. 2014;95(5):550–7.CrossRefPubMed
13.
go back to reference Han B, Mao J, Chien JY, Hall SD. Optimization of drug–drug interaction study design: comparison of minimal physiologically based pharmacokinetic models on prediction of CYP3A inhibition by ketoconazole. Drug Metab Dispos. 2013;41(7):1329–38.CrossRefPubMed Han B, Mao J, Chien JY, Hall SD. Optimization of drug–drug interaction study design: comparison of minimal physiologically based pharmacokinetic models on prediction of CYP3A inhibition by ketoconazole. Drug Metab Dispos. 2013;41(7):1329–38.CrossRefPubMed
14.
go back to reference Einolf HJ. Comparison of different approaches to predict metabolic drug–drug interactions. Xenobiotica. 2007;37(10):1257–94.CrossRefPubMed Einolf HJ. Comparison of different approaches to predict metabolic drug–drug interactions. Xenobiotica. 2007;37(10):1257–94.CrossRefPubMed
15.
go back to reference Fahmi OA, Hurst S, Plowchalk D, Cook J, Guo F, Youdim K, et al. Comparison of different algorithms for predicting clinical drug–drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds as precipitants of interaction. Drug Metab Dispos. 2009;37(8):1658–66.CrossRefPubMed Fahmi OA, Hurst S, Plowchalk D, Cook J, Guo F, Youdim K, et al. Comparison of different algorithms for predicting clinical drug–drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds as precipitants of interaction. Drug Metab Dispos. 2009;37(8):1658–66.CrossRefPubMed
16.
go back to reference Varma MV, Lin J, Bi YA, Kimoto E, Rodrigues AD. Quantitative rationalization of Gemfibrozil drug interactions: consideration of transporters-enzyme interplay and the role of circulating metabolite Gemfibrozil 1-O-beta-glucuronide. Drug Metab Dispos. 2015;43(7):1108–18.CrossRefPubMed Varma MV, Lin J, Bi YA, Kimoto E, Rodrigues AD. Quantitative rationalization of Gemfibrozil drug interactions: consideration of transporters-enzyme interplay and the role of circulating metabolite Gemfibrozil 1-O-beta-glucuronide. Drug Metab Dispos. 2015;43(7):1108–18.CrossRefPubMed
17.
go back to reference Huang SM, Abernethy DR, Wang Y, Zhao P, Zineh I. The utility of modeling and simulation in drug development and regulatory review. J Pharm Sci. 2013;102(9):2912–23.CrossRefPubMed Huang SM, Abernethy DR, Wang Y, Zhao P, Zineh I. The utility of modeling and simulation in drug development and regulatory review. J Pharm Sci. 2013;102(9):2912–23.CrossRefPubMed
18.
go back to reference Snoeys J, Beumont M, Monshouwer M, Ouwerkerk-Mahadevan S. Mechanistic understanding of the nonlinear pharmacokinetics and intersubject variability of simeprevir: a PBPK-guided drug development approach. Clin Pharmacol Ther. 2016;99(2):224–34.CrossRefPubMed Snoeys J, Beumont M, Monshouwer M, Ouwerkerk-Mahadevan S. Mechanistic understanding of the nonlinear pharmacokinetics and intersubject variability of simeprevir: a PBPK-guided drug development approach. Clin Pharmacol Ther. 2016;99(2):224–34.CrossRefPubMed
19.
go back to reference Guo H, Liu C, Li J, Zhang M, Hu M, Xu P, et al. A mechanistic physiologically based pharmacokinetic-enzyme turnover model involving both intestine and liver to predict CYP3A induction-mediated drug–drug interactions. J Pharm Sci. 2013;102(8):2819–36.CrossRefPubMed Guo H, Liu C, Li J, Zhang M, Hu M, Xu P, et al. A mechanistic physiologically based pharmacokinetic-enzyme turnover model involving both intestine and liver to predict CYP3A induction-mediated drug–drug interactions. J Pharm Sci. 2013;102(8):2819–36.CrossRefPubMed
20.
go back to reference Vieira ML, Kirby B, Ragueneau-Majlessi I, Galetin A, Chien JY, Einolf HJ, et al. Evaluation of various static in vitro-in vivo extrapolation models for risk assessment of the CYP3A inhibition potential of an investigational drug. Clin Pharmacol Ther. 2014;95(2):189–98.CrossRefPubMed Vieira ML, Kirby B, Ragueneau-Majlessi I, Galetin A, Chien JY, Einolf HJ, et al. Evaluation of various static in vitro-in vivo extrapolation models for risk assessment of the CYP3A inhibition potential of an investigational drug. Clin Pharmacol Ther. 2014;95(2):189–98.CrossRefPubMed
21.
go back to reference Guest EJ, Rowland-Yeo K, Rostami-Hodjegan A, Tucker GT, Houston JB, Galetin A. Assessment of algorithms for predicting drug–drug interactions via inhibition mechanisms: comparison of dynamic and static models. Br J Clin Pharmacol. 2011;71(1):72–87.CrossRefPubMedPubMedCentral Guest EJ, Rowland-Yeo K, Rostami-Hodjegan A, Tucker GT, Houston JB, Galetin A. Assessment of algorithms for predicting drug–drug interactions via inhibition mechanisms: comparison of dynamic and static models. Br J Clin Pharmacol. 2011;71(1):72–87.CrossRefPubMedPubMedCentral
Metadata
Title
Predictive Performance of Physiologically-Based Pharmacokinetic Models in Predicting Drug–Drug Interactions Involving Enzyme Modulation
Authors
Chia-Hsiang Hsueh
Vicky Hsu
Yuzhuo Pan
Ping Zhao
Publication date
01-10-2018
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 10/2018
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-018-0635-8

Other articles of this Issue 10/2018

Clinical Pharmacokinetics 10/2018 Go to the issue