Skip to main content
Top
Published in: Clinical Pharmacokinetics 11/2019

Open Access 01-11-2019 | Current Opinion

Requirements to Establishing Confidence in Physiologically Based Pharmacokinetic (PBPK) Models and Overcoming Some of the Challenges to Meeting Them

Authors: Sheila Annie Peters, Hugues Dolgos

Published in: Clinical Pharmacokinetics | Issue 11/2019

Login to get access

Abstract

When scientifically well-founded, the mechanistic basis of physiologically based pharmacokinetic (PBPK) models can help reduce the uncertainty and increase confidence in extrapolations outside the studied scenarios or studied populations. However, it is not always possible to establish mechanistically credible PBPK models. Requirements to establishing confidence in PBPK models, and challenges to meeting these requirements, are presented in this article. Parameter non-identifiability is the most challenging among the barriers to establishing confidence in PBPK models. Using case examples of small molecule drugs, this article examines the use of hypothesis testing to overcome parameter non-identifiability issues, with the objective of enhancing confidence in the mechanistic basis of PBPK models and thereby improving the quality of predictions that are meant for internal decisions and regulatory submissions. When the mechanistic basis of a PBPK model cannot be established, we propose the use of simpler models or evidence-based approaches.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jones HM, et al. Physiologically based pharmacokinetic modeling in drug discovery and development: a pharmaceutical industry perspective. Clin Pharmacol Ther. 2015;97:247–62.PubMed Jones HM, et al. Physiologically based pharmacokinetic modeling in drug discovery and development: a pharmaceutical industry perspective. Clin Pharmacol Ther. 2015;97:247–62.PubMed
2.
go back to reference Peters SA. Physiologically-based pharmacokinetic (PBPK) modelling and simulations: principles, methods and applications in the pharmaceutical industry. Hoboken: Wiley; 2012. Peters SA. Physiologically-based pharmacokinetic (PBPK) modelling and simulations: principles, methods and applications in the pharmaceutical industry. Hoboken: Wiley; 2012.
3.
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;102:2912–23.PubMed 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;102:2912–23.PubMed
4.
5.
go back to reference Luzon E, Blake K, Cole S, Nordmark A, Versantvoort C, Berglund EG. Physiologically based pharmacokinetic modeling in regulatory decision-making at the European Medicines Agency. Clin Pharmacol Ther. 2017;102(1):98–105.PubMed Luzon E, Blake K, Cole S, Nordmark A, Versantvoort C, Berglund EG. Physiologically based pharmacokinetic modeling in regulatory decision-making at the European Medicines Agency. Clin Pharmacol Ther. 2017;102(1):98–105.PubMed
6.
go back to reference Sato M, et al. Quantitative modeling and simulation in PMDA: a Japanese regulatory perspective. CPT Pharmacomet Syst Pharmacol. 2017;6:413–5. Sato M, et al. Quantitative modeling and simulation in PMDA: a Japanese regulatory perspective. CPT Pharmacomet Syst Pharmacol. 2017;6:413–5.
7.
go back to reference Shebley M, Sandhu P, Emami Riedmaier A, Jamei M, Narayanan R, Patel A, et al. Physiologically-based pharmacokinetic model qualification and reporting. Clin Pharmacol Ther. 2018;104:88–110.PubMedPubMedCentral Shebley M, Sandhu P, Emami Riedmaier A, Jamei M, Narayanan R, Patel A, et al. Physiologically-based pharmacokinetic model qualification and reporting. Clin Pharmacol Ther. 2018;104:88–110.PubMedPubMedCentral
8.
go back to reference Peters SA. Evaluation of a generic physiologically based pharmacokinetic model for line shape analysis. Clin Pharmacokinet. 2008;47:261–75.PubMed Peters SA. Evaluation of a generic physiologically based pharmacokinetic model for line shape analysis. Clin Pharmacokinet. 2008;47:261–75.PubMed
9.
go back to reference Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73.PubMed Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73.PubMed
10.
go back to reference Grimstein M, Yang Y, Zhang X, Grillo J, Huang SM, Zineh I, et al. Physiologically-based pharmacokinetic (PBPK) modeling in regulatory science: an update from the US Food and Drug Administration’s Office of Clinical Pharmacology. J Pharm Sci. 2019;108(1):21–5.PubMed Grimstein M, Yang Y, Zhang X, Grillo J, Huang SM, Zineh I, et al. Physiologically-based pharmacokinetic (PBPK) modeling in regulatory science: an update from the US Food and Drug Administration’s Office of Clinical Pharmacology. J Pharm Sci. 2019;108(1):21–5.PubMed
11.
go back to reference Wang Y, Huang SM. Commentary on fit-for-purpose models for regulatory applications. J Pharm Sci. 2019;108(1):18–20.PubMed Wang Y, Huang SM. Commentary on fit-for-purpose models for regulatory applications. J Pharm Sci. 2019;108(1):18–20.PubMed
14.
go back to reference Mehrotra N, Bhattaram A, Earp JC, Florian J, Krudys K, Lee JE, et al. Role of quantitative clinical pharmacology in pediatric approval and labeling. Drug Metab Dispos. 2016;44(7):924–33.PubMed Mehrotra N, Bhattaram A, Earp JC, Florian J, Krudys K, Lee JE, et al. Role of quantitative clinical pharmacology in pediatric approval and labeling. Drug Metab Dispos. 2016;44(7):924–33.PubMed
16.
go back to reference Manolis E, Rohou S, Hemmings R, Salmonson T, Karlsson M, Milligan PA. The role of modeling and simulation in development and registration of medicinal products: output from the EFPIA/EMA modeling and simulation workshop. CPT Pharmacomet Syst Pharmacol. 2013;2:e31. Manolis E, Rohou S, Hemmings R, Salmonson T, Karlsson M, Milligan PA. The role of modeling and simulation in development and registration of medicinal products: output from the EFPIA/EMA modeling and simulation workshop. CPT Pharmacomet Syst Pharmacol. 2013;2:e31.
17.
go back to reference Abduljalil K, Cain T, Humphries H, Rostami-Hodjegan A. Deciding on success criteria for predictability of pharmacokinetic parameters from in vitro studies: an analysis based on in vivo observations. Drug Metab Dispos. 2014;42:1478–84.PubMed Abduljalil K, Cain T, Humphries H, Rostami-Hodjegan A. Deciding on success criteria for predictability of pharmacokinetic parameters from in vitro studies: an analysis based on in vivo observations. Drug Metab Dispos. 2014;42:1478–84.PubMed
18.
go back to reference Rostami-Hodjegan A. Reverse translation in PBPK and QSP: going backwards in order to go forward with confidence. Clin Pharmacol Ther. 2018;103:224–32.PubMed Rostami-Hodjegan A. Reverse translation in PBPK and QSP: going backwards in order to go forward with confidence. Clin Pharmacol Ther. 2018;103:224–32.PubMed
19.
go back to reference Yau E, Petersson C, Dolgos H, Peters SA. A comparative evaluation of models to predict human intestinal metabolism from nonclinical data. Biopharm Drug Dispos. 2017;38(3):163–86.PubMedPubMedCentral Yau E, Petersson C, Dolgos H, Peters SA. A comparative evaluation of models to predict human intestinal metabolism from nonclinical data. Biopharm Drug Dispos. 2017;38(3):163–86.PubMedPubMedCentral
20.
go back to reference Johnson TN, Zhou D, Bui KH. Development of physiologically based pharmacokinetic model to evaluate the relative systemic exposure to quetiapine after administration of IR and XR formulations to adults, children and adolescents. Biopharm Drug Dispos. 2014;35:341–52.PubMed Johnson TN, Zhou D, Bui KH. Development of physiologically based pharmacokinetic model to evaluate the relative systemic exposure to quetiapine after administration of IR and XR formulations to adults, children and adolescents. Biopharm Drug Dispos. 2014;35:341–52.PubMed
21.
go back to reference Hansmann S, Darwich A, Margolskee A, Aarons L, Dressman J. Forecasting oral absorption across biopharmaceutics classification system classes with physiologically based pharmacokinetic models. J Pharm Pharmacol. 2016;68(12):1501–15.PubMed Hansmann S, Darwich A, Margolskee A, Aarons L, Dressman J. Forecasting oral absorption across biopharmaceutics classification system classes with physiologically based pharmacokinetic models. J Pharm Pharmacol. 2016;68(12):1501–15.PubMed
22.
go back to reference Dodd S, Kollipara S, Sanchez-Felix M, Kim H, Meng Q, Beato S, et al. Prediction of ARA/PPI drug–drug interactions at the drug discovery and development interface. J Pharm Sci. 2019;108(1):87–101.PubMed Dodd S, Kollipara S, Sanchez-Felix M, Kim H, Meng Q, Beato S, et al. Prediction of ARA/PPI drug–drug interactions at the drug discovery and development interface. J Pharm Sci. 2019;108(1):87–101.PubMed
23.
go back to reference Samant T, et al. Ribociclib bioavailability is not affected by gastric pH changes or food intake: in silico and clinical evaluations. Clin Pharmacol Ther. 2018;104(2):374–83.PubMed Samant T, et al. Ribociclib bioavailability is not affected by gastric pH changes or food intake: in silico and clinical evaluations. Clin Pharmacol Ther. 2018;104(2):374–83.PubMed
24.
go back to reference Tistaert C, Heimbach T, Xia B, Parrott N, Samant TS, Kesisoglou F. Food effect projections via physiologically based pharmacokinetic modeling: predictive case studies. J Pharm Sci. 2019;108(1):592–602.PubMed Tistaert C, Heimbach T, Xia B, Parrott N, Samant TS, Kesisoglou F. Food effect projections via physiologically based pharmacokinetic modeling: predictive case studies. J Pharm Sci. 2019;108(1):592–602.PubMed
25.
go back to reference Rekic D, Roshammar D, Mukonzo J, Ashton M. In silico prediction of efavirenz and rifampicin drug–drug interaction considering weight and CYP2B6 phenotype. Br J Clin Pharmacol. 2011;71:536–43.PubMedPubMedCentral Rekic D, Roshammar D, Mukonzo J, Ashton M. In silico prediction of efavirenz and rifampicin drug–drug interaction considering weight and CYP2B6 phenotype. Br J Clin Pharmacol. 2011;71:536–43.PubMedPubMedCentral
26.
go back to reference Mao J, Johnson TR, Shen Z, Yamazaki S. Prediction of crizotinib-midazolam interaction using the Simcyp population-based simulator: comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos. 2013;41:343–52.PubMed Mao J, Johnson TR, Shen Z, Yamazaki S. Prediction of crizotinib-midazolam interaction using the Simcyp population-based simulator: comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos. 2013;41:343–52.PubMed
27.
go back to reference Yamazaki S, Johnson TR, Smith BJ. Prediction of drug–drug interactions with crizotinib as the CYP3A substrate using a physiologically based pharmacokinetic model. Drug Metab Dispos. 2015;43:1417–29.PubMed Yamazaki S, Johnson TR, Smith BJ. Prediction of drug–drug interactions with crizotinib as the CYP3A substrate using a physiologically based pharmacokinetic model. Drug Metab Dispos. 2015;43:1417–29.PubMed
28.
go back to reference Arya V, Zhao P, Reynolds KS, Mishra P, Younis IR. Utilizing PBPK modeling to evaluate the potential of a significant drug–drug interaction between clopidogrel and dasabuvir: a scientific perspective. Clin Pharmacol Ther. 2017;102(4):578–80.PubMed Arya V, Zhao P, Reynolds KS, Mishra P, Younis IR. Utilizing PBPK modeling to evaluate the potential of a significant drug–drug interaction between clopidogrel and dasabuvir: a scientific perspective. Clin Pharmacol Ther. 2017;102(4):578–80.PubMed
29.
go back to reference Shebley M, Fu W, Badri P, Bow D, Fischer V. Physiologically based pharmacokinetic modeling suggests limited drug–drug interaction between clopidogrel and dasabuvir. Clin Pharmacol Ther. 2017;102(4):679–87.PubMedPubMedCentral Shebley M, Fu W, Badri P, Bow D, Fischer V. Physiologically based pharmacokinetic modeling suggests limited drug–drug interaction between clopidogrel and dasabuvir. Clin Pharmacol Ther. 2017;102(4):679–87.PubMedPubMedCentral
30.
go back to reference Einolf HJ, et al. Physiologically-based pharmacokinetic model predictions of panobinostat (LBH589) as a victim and perpetrator of drug–drug interactions. Drug Metab Dispos. 2017;45(12):1304–16.PubMed Einolf HJ, et al. Physiologically-based pharmacokinetic model predictions of panobinostat (LBH589) as a victim and perpetrator of drug–drug interactions. Drug Metab Dispos. 2017;45(12):1304–16.PubMed
31.
go back to reference Rose RH, Neuhoff S, Abduljalil K, Chetty M, Rostami-Hodjegan A, Jamei M. Application of a physiologically based pharmacokinetic model to predict OATP1B1-related variability in pharmacodynamics of rosuvastatin. CPT Pharmacomet Syst Pharmacol. 2014;3:e124. Rose RH, Neuhoff S, Abduljalil K, Chetty M, Rostami-Hodjegan A, Jamei M. Application of a physiologically based pharmacokinetic model to predict OATP1B1-related variability in pharmacodynamics of rosuvastatin. CPT Pharmacomet Syst Pharmacol. 2014;3:e124.
32.
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.PubMed 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.PubMed
33.
go back to reference Shebley M, Liu J, Kavetskaia O, Sydor J, de Morais SM, Fischer V, et al. Mechanisms and predictions of drug–drug interactions of the hepatitis c virus three direct-acting antiviral regimen: paritaprevir/ritonavir, ombitasvir, and dasabuvir. Drug Metab Dispos. 2017;45(7):755–64.PubMed Shebley M, Liu J, Kavetskaia O, Sydor J, de Morais SM, Fischer V, et al. Mechanisms and predictions of drug–drug interactions of the hepatitis c virus three direct-acting antiviral regimen: paritaprevir/ritonavir, ombitasvir, and dasabuvir. Drug Metab Dispos. 2017;45(7):755–64.PubMed
34.
go back to reference Mao J, Doshi U, Wright M, Hop CECA, Li AP, Chen Y. Prediction of the pharmacokinetics of pravastatin as an OATP substrate using plateable human hepatocytes with human plasma data and PBPK modeling. CPT Pharmacomet Syst Pharmacol. 2018;7(4):251–8. Mao J, Doshi U, Wright M, Hop CECA, Li AP, Chen Y. Prediction of the pharmacokinetics of pravastatin as an OATP substrate using plateable human hepatocytes with human plasma data and PBPK modeling. CPT Pharmacomet Syst Pharmacol. 2018;7(4):251–8.
35.
go back to reference Chen Y, Zhu R, Ma F, Mao J, Chen EC, Choo EF, et al. Assessment of OATP transporter-mediated drug–drug interaction using physiologically-based pharmacokinetic (PBPK) modeling—a case example. Biopharm Drug Dispos. 2018;39(9):420–30.PubMed Chen Y, Zhu R, Ma F, Mao J, Chen EC, Choo EF, et al. Assessment of OATP transporter-mediated drug–drug interaction using physiologically-based pharmacokinetic (PBPK) modeling—a case example. Biopharm Drug Dispos. 2018;39(9):420–30.PubMed
36.
go back to reference Graham L, Lloyd S. Biomedical accelerator mass spectrometry: recent applications in metabolism and pharmacokinetics. Expert Opin Drug Metab Toxicol. 2008;4(8):1021–33. Graham L, Lloyd S. Biomedical accelerator mass spectrometry: recent applications in metabolism and pharmacokinetics. Expert Opin Drug Metab Toxicol. 2008;4(8):1021–33.
37.
go back to reference Penner N, Klunk LJ, Prakash C. Human radiolabeled mass balance studies: objectives, utilities and limitations. Biopharm Drug Dispos. 2009;30:185–203.PubMed Penner N, Klunk LJ, Prakash C. Human radiolabeled mass balance studies: objectives, utilities and limitations. Biopharm Drug Dispos. 2009;30:185–203.PubMed
38.
go back to reference Tsamandouras N, Rostami-Hodjegan A, Aarons L. Combining the ‘bottom up’ and ‘top down’ approaches in pharmacokinetic modelling: fitting PBPK models to observed clinical data. Br J Clin Pharmacol. 2015;79(1):48–55.PubMed Tsamandouras N, Rostami-Hodjegan A, Aarons L. Combining the ‘bottom up’ and ‘top down’ approaches in pharmacokinetic modelling: fitting PBPK models to observed clinical data. Br J Clin Pharmacol. 2015;79(1):48–55.PubMed
39.
go back to reference Cubitt HE, Yeo KR, Howgate EM, Rostami-Hodjegan A, Barter ZE. Sources of interindividual variability in IVIVE of clearance: an investigation into the prediction of benzodiazepine clearance using a mechanistic population-based pharmacokinetic model. Xenobiotica. 2011;41(8):623–38.PubMed Cubitt HE, Yeo KR, Howgate EM, Rostami-Hodjegan A, Barter ZE. Sources of interindividual variability in IVIVE of clearance: an investigation into the prediction of benzodiazepine clearance using a mechanistic population-based pharmacokinetic model. Xenobiotica. 2011;41(8):623–38.PubMed
40.
go back to reference McNally K, Cotton R, Loizou GD. Front Pharmacol. A Workflow for Global Sensitivity Analysis of PBPK Models. 2011;23:2–31. McNally K, Cotton R, Loizou GD. Front Pharmacol. A Workflow for Global Sensitivity Analysis of PBPK Models. 2011;23:2–31.
41.
go back to reference Peters SA, Hultin L. Early identification of drug-induced impairment of gastric emptying through physiologically based pharmacokinetic (PBPK) simulation of plasma concentration-time profiles in rat. J Pharmacokinet Pharmacodyn. 2008;35:1–30.PubMed Peters SA, Hultin L. Early identification of drug-induced impairment of gastric emptying through physiologically based pharmacokinetic (PBPK) simulation of plasma concentration-time profiles in rat. J Pharmacokinet Pharmacodyn. 2008;35:1–30.PubMed
42.
go back to reference Peters SA. Identification of intestinal loss of a drug through physiologically based pharmacokinetic simulation of plasma concentration-time profiles. Clin Pharmacokinet. 2008;47:245–59.PubMed Peters SA. Identification of intestinal loss of a drug through physiologically based pharmacokinetic simulation of plasma concentration-time profiles. Clin Pharmacokinet. 2008;47:245–59.PubMed
43.
go back to reference EFPIA MID3 Workgroup, Marshall SF, Burghaus R, Cosson V, Cheung SYA, Chenel M, et al. Good practices in model-informed drug discovery and development: practice, application, and documentation. CPT Pharmacomet Syst Pharmacol. 2016;5(3):93–122. EFPIA MID3 Workgroup, Marshall SF, Burghaus R, Cosson V, Cheung SYA, Chenel M, et al. Good practices in model-informed drug discovery and development: practice, application, and documentation. CPT Pharmacomet Syst Pharmacol. 2016;5(3):93–122.
44.
go back to reference Parkman HP, Urbain JL, Knight LC, Brown KL, Trate DM, Miller MA, et al. Effect of gastric acid suppressants on human gastric motility. Gut. 1998;42(2):243–50.PubMedPubMedCentral Parkman HP, Urbain JL, Knight LC, Brown KL, Trate DM, Miller MA, et al. Effect of gastric acid suppressants on human gastric motility. Gut. 1998;42(2):243–50.PubMedPubMedCentral
Metadata
Title
Requirements to Establishing Confidence in Physiologically Based Pharmacokinetic (PBPK) Models and Overcoming Some of the Challenges to Meeting Them
Authors
Sheila Annie Peters
Hugues Dolgos
Publication date
01-11-2019
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 11/2019
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-019-00790-0

Other articles of this Issue 11/2019

Clinical Pharmacokinetics 11/2019 Go to the issue