Skip to main content
Log in

Physiologically-Based Pharmacokinetic Modelling of Entrectinib Parent and Active Metabolite to Support Regulatory Decision-Making

  • Original Research Article
  • Published:
European Journal of Drug Metabolism and Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objective

Entrectinib is a selective inhibitor of ROS1/TRK/ALK kinases, recently approved for oncology indications. Entrectinib is predominantly cleared by cytochrome P450 (CYP) 3A4, and modulation of CYP3A enzyme activity profoundly alters the pharmacokinetics of both entrectinib and its active metabolite M5. We describe development of a combined physiologically based pharmacokinetic (PBPK) model for entrectinib and M5 to support dosing recommendations when entrectinib is co-administered with CYP3A4 inhibitors or inducers.

Methods

A PBPK model was established in Simcyp® Simulator. The initial model based on in vitro–in vivo extrapolation was refined using sensitivity analysis and non-linear mixed effects modeling to optimize parameter estimates and to improve model fit to data from a clinical drug–drug interaction study with the strong CYP3A4 inhibitor, itraconazole. The model was subsequently qualified against clinical data, and the final qualified model used to simulate the effects of moderate to strong CYP3A4 inhibitors and inducers on entrectinib and M5 pharmacokinetics.

Results

The final model showed good predictive performance for entrectinib and M5, meeting commonly used predictive performance acceptance criteria in each case. The model predicted that co-administration of various moderate CYP3A4 inhibitors (verapamil, erythromycin, clarithromycin, fluconazole, and diltiazem) would result in an average increase in entrectinib exposure between 2.2- and 3.1-fold, with corresponding average increases for M5 of approximately 2-fold. Co-administration of moderate CYP3A4 inducers (efavirenz, carbamazepine, phenytoin) was predicted to result in an average decrease in entrectinib exposure between 45 and 79%, with corresponding average decreases for M5 of approximately 50%.

Conclusions

The model simulations were used to derive dosing recommendations for co-administering entrectinib with CYP3A4 inhibitors or inducers. PBPK modeling has been used in lieu of clinical studies to enable regulatory decision-making.

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
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Jones H, Rowland-Yeo K. Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT Pharmacometrics Syst Pharmacol. 2013;2:e63.

    Article  Google Scholar 

  2. Jones HM, Chen Y, Gibson C, Heimbach T, Parrott N, Peters SA, Snoeys J, Upreti VV, Zheng M, Hall SD. Physiologically based pharmacokinetic modeling in drug discovery and development: a pharmaceutical industry perspective. Clin Pharmacol Ther. 2015;97(3):247–62.

    Article  CAS  Google Scholar 

  3. Jamei M. Recent advances in development and application of physiologically-based pharmacokinetic (pbpk) models: a transition from academic curiosity to regulatory acceptance. Curr Pharmacol Rep. 2016;2:161–9.

    Article  CAS  Google Scholar 

  4. Templeton IE, Chen Y, Mao J, Lin J, Yu H, Peters S, Shebley M, Varma MV. Quantitative prediction of drug-drug interactions involving inhibitory metabolites in drug development: how can physiologically based pharmacokinetic modeling help? CPT Pharmacomet Syst Pharmacol. 2016;5(10):505–15.

    Article  CAS  Google Scholar 

  5. Zhuang X, Lu C. PBPK modeling and simulation in drug research and development. Acta Pharm Sin B. 2016;6(5):430–40.

    Article  Google Scholar 

  6. Miller NA, Reddy MB, Heikkinen AT, Lukacova V, Parrott N. Physiologically based pharmacokinetic modelling for first-in-human predictions: an updated model building strategy illustrated with challenging industry case studies. Clin Pharmacokinet. 2019;58(6):727–46.

    Article  CAS  Google Scholar 

  7. Peters SA, Dolgos H. Requirements to establishing confidence in physiologically based pharmacokinetic (PBPK) models and overcoming some of the challenges to meeting them. Clin Pharmacokinet. 2019;58(11):1355–71.

    Article  CAS  Google Scholar 

  8. Taskar KS, Pilla Reddy V, Burt H, et al. Physiologically-based pharmacokinetic models for evaluating membrane transporter mediated drug-drug interactions: current capabilities, case studies, future opportunities, and recommendations. Clin Pharmacol Ther. 2020;107(5):1082–115. https://doi.org/10.1002/cpt.1693.

    Article  PubMed  Google Scholar 

  9. Zhao P, Rowland M, Huang SM. Best practice in the use of physiologically based pharmacokinetic modeling and simulation to address clinical pharmacology regulatory questions. Clin Pharmacol Ther. 2012;92(1):17–20.

    Article  CAS  Google Scholar 

  10. Rowland M, Lesko LJ, Rostami-Hodjegan A. Physiologically based pharmacokinetics is impacting drug development and regulatory decision Making. CPT Pharmacomet Syst Pharmacol. 2015;4(6):313–5.

    Article  CAS  Google Scholar 

  11. Wagner C, Pan Y, Hsu VJA, 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):117–112.

    Article  CAS  Google Scholar 

  12. Shebley M, Sandhu P, Emami Riedmaier AM, et al. Physiologically based pharmacokinetic model qualification and reporting procedures for regulatory submissions: a consortium perspective. Clin Pharmacol Ther. 2018;104(1):88–110.

    Article  Google Scholar 

  13. Grimstein M, Yang Y, Zhang XJ, et al. Physiologically based pharmacokinetic modeling in regulatory science: an update from the U.S. Food and Drug Administration’s Office of Clinical Pharmacology. J Pharm Sci. 2019;108(1):21–5.

    Article  CAS  Google Scholar 

  14. de Zwart L, Snoeys J, De Jong JJ, et al. Ibrutinib dosing strategies based on interaction potential of CYP3A4 perpetrators using physiologically based pharmacokinetic modeling. Clin Pharmacol Ther. 2016;100(5):548–57.

    Article  Google Scholar 

  15. Cleary Y, Gertz M, Djebli N, et al. Mechanistic Modeling approaches to simultaneously estimate FMCYP3A and FG of CYP3A substrates from clinical DDI study data. Clinical Pharmacology & Therapeutics. 2020;107(S1);S27.

  16. Meneses-Lorente G, Bentley D, Guerini E, et al. Characterization of the pharmacokinetics of entrectinib and its active m5 metabolite in healthy volunteers and patients with solid tumors. Clin Pharmacol Ther. 2020;107(S1):S27.

    Google Scholar 

  17. Pilla Reddy V, Bui K, Scarfe GD, et al. Physiologically based pharmacokinetic modeling for olaparib dosing recommendations: bridging formulations, drug interactions, and patient populations. Clin Pharmacol Ther. 2019;105(1):229–41.

    Article  CAS  Google Scholar 

  18. Umehara K, Huth F, Jin YH, et al. Drug-drug interaction (DDI) assessments of ruxolitinib, a dual substrate of CYP3A4 and CYP2C9, using a verified physiologically based pharmacokinetic (PBPK) model to support regulatory submissions. Drug Metab Pers Ther. 2019. https://doi.org/10.1515/dmpt-2018-0042.

  19. Rowland Yeo K, Jamei M, Yang JGT, et al. Physiologically based mechanistic modelling to predict complex drug-drug interactions involving simultaneous competitive and time-dependent enzyme inhibition by parent compound and its metabolite in both liver and gut - the effect of diltiazem on the time-course of exposure to triazolam. Eur J Pharm Sci. 2010;39(5):298–309.

    Article  CAS  Google Scholar 

  20. Guo J, Zhou D, Li Y, Khanh BH. Physiologically based pharmacokinetic modeling to predict complex drug-drug interactions: a case study of AZD2327 and its metabolite, competitive and time-dependent CYP3A inhibitors. Biopharm Drug Dispos. 2015;36(8):507–19.

    Article  CAS  Google Scholar 

  21. Asaumi R, Toshimoto K, Tobe YK, et al. Comprehensive PBPK model of rifampicin for quantitative prediction of complex drug-drug interactions: CYP3A/2C9 induction and OATP inhibition effects. CPT Pharmacomet Syst Pharmacol. 2018;7(3):186–96.

    Article  CAS  Google Scholar 

  22. Yoshida K, Maeda K, Konagaya A, et al. Accurate estimation of in vivo inhibition constants of inhibitors and fraction metabolized of substrates with physiologically based pharmacokinetic drug-drug interaction models incorporating parent drugs and metabolites of substrates with cluster newton method. Drug Metab Dispos. 2018;46(11):1805–16.

    Article  CAS  Google Scholar 

  23. Marsousi N, Samer CF, Fontana P, et al. Coadministration of ticagrelor and ritonavir: toward prospective dose adjustment to maintain an optimal platelet inhibition using the PBPK approach. Clin Pharmacol Ther. 2016;100(3):295–304.

    Article  CAS  Google Scholar 

  24. Cleary Y, Gertz M, Morcos PNL, et al. Model-based assessments of CYP-mediated drug-drug interaction risk of alectinib: physiologically based pharmacokinetic modeling supported clinical development. Clin Pharmacol Ther. 2018;104(3):505–14.

    Article  CAS  Google Scholar 

  25. Gu H, Dutreix S, Rebello ST, et al. Simultaneous physiologically based pharmacokinetic (PBPK) modeling of parent and active metabolites to investigate complex CYP3A4 drug-drug interaction potential: a case example of midostaurin. Drug Metab Dispos. 2018;46(2):109–21.

    Article  CAS  Google Scholar 

  26. Zhou D, Podoll T, Xu YG, et al. Evaluation of the drug-drug interaction potential of acalabrutinib and its active metabolite, ACP-5862, using a physiologically-based pharmacokinetic modeling approach. CPT Pharmacometrics Syst Pharmacol. 2019;8(7):489–99.

    Article  CAS  Google Scholar 

  27. Posada MM, Morse BL, Turner PK, Kulanthaivel P, Hall SD, Dickinson GL. Predicting clinical effects of CYP3A4 modulators on abemaciclib and active metabolites exposure using physiologically based pharmacokinetic modeling [published online ahead of print, 2020 Feb 20]. J Clin Pharmacol. 2020. https://doi.org/10.1002/jcph.1584.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Van den Bergh A, Snoeys J, De Zwart L, et al. Pharmacokinetic drug-drug interaction of apalutamide, part 2: investigating interaction potential using a physiologically based pharmacokinetic [published online ahead of print, 2020 Apr 27]. Clin Pharmacokinet. 2020. https://doi.org/10.1007/s40262-020-00881-3.

    Article  PubMed  Google Scholar 

  29. Li R, Barton HA, Yates PD, Ghosh A, Wolford AC, Riccardi KA, Maurer TS. A ‘“middle-out”’ approach to human pharmacokinetic predictions for OATP substrates using physiologically-based pharmacokinetic modeling. J Pharmacokinet Pharmacodyn. 2014;41:197–209.

    Article  CAS  Google Scholar 

  30. 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.

    Article  CAS  Google Scholar 

  31. Tylutki Z, Polak S, Wiśniowska B. Top-down, bottom-up and middle-out strategies for drug cardiac safety assessment via modeling and simulations. Curr Pharmacol Rep. 2016;2(4):171–7.

    Article  CAS  Google Scholar 

  32. Rostami-Hodjegan A. Reverse translation in PBPK and QSP: going backwards in order to go forward with confidence. Clin Pharmacol Ther. 2018;103(2):224–32.

    Article  Google Scholar 

  33. Drilon A, Siena S, Ou SI, et al. Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK Inhibitor entrectinib: combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017;7(4):400–9. https://doi.org/10.1158/2159-8290.CD-16-1237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Meneses-Lorente G, Fowler S, Guerini E, et al. In vitro and clinical investigations to determine the drug-drug interaction potential of entrectinib. In: Under Press in Investigational New Drugs.

  35. Rodgers T, Rowland M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions [published correction appears in J Pharm Sci. 2007 Nov;96(11):3153-4]. J Pharm Sci. 2006;95(6):1238–57. https://doi.org/10.1002/jps.20502.

    Article  CAS  PubMed  Google Scholar 

  36. Parrott N, Stillhart C, Lindenberg M, et al. Physiologically based absorption modelling to explore the impact of food and gastric pH changes on the pharmacokinetics of entrectinib. AAPS J. 2020;22(4):78. https://doi.org/10.1208/s12248-020-00463-y.

    Article  CAS  PubMed  Google Scholar 

  37. Jamei M, Turner D, Yang J, et al. Population-based mechanistic prediction of oral drug absorption. AAPS J. 2009;11(2):225–37. https://doi.org/10.1208/s12248-009-9099-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Meneses-Lorente G, Fowler S, Guerini E, et al. In vitro and clinical investigations to determine the drug-drug interaction potential of entrectinib, a small molecule inhibitor of neurotrophic tyrosine receptor kinase (NTRK). Invest New Drugs. 2021. https://doi.org/10.1007/s10637-021-01156-9.

    Article  PubMed  PubMed Central  Google Scholar 

  39. 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. https://doi.org/10.1124/dmd.115.065920.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Jamei M, Dickinson GL, Rostami-Hodjegan A. A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: A tale of “bottom-up” vs “top-down” recognition of covariates [published correction appears in Drug Metab Pharmacokinet. 2009;24(5):488]. Drug Metab Pharmacokinet. 2009;24(1):53–75. https://doi.org/10.2133/dmpk.24.53.

    Article  CAS  PubMed  Google Scholar 

  41. Mercier F, Djebli N, González-Sales M, Meneses-Lorente G, Jaminion F, Phipps A, Frey N. Exposure–response analysis of entrectinib supports the recommended dose in patients with advanced/metastatic solid tumors. Clinical Pharmacology & Therapeutics; Vol 107 Supplement S1;S50

  42. Drilon A, Siena S, Dziadziuszko R, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21(2):261–70.

    Article  CAS  Google Scholar 

  43. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020;21(2):271–82.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The clinical studies reported in this manuscript were funded by F. Hoffmann-La-Roche (formerly Ignyta Inc., a member of the Roche Group). The modeling analyses were also funded by F. Hoffmann-La-Roche.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nassim Djebli.

Ethics declarations

Funding

The clinical studies reported in this manuscript were funded by F. Hoffmann-La-Roche (formerly Ignyta Inc., a member of the Roche Group). The modeling analyses were also funded by F. Hoffmann-La-Roche. Administrative support was provided by Ashfield Medcomms, an Ashfield Health company, and was funded by F. Hoffmann-La Roche Ltd.

Conflicts of Interest

G.M-L. is an employee of Roche Products Ltd. N.D. E.G. Y.C., and A.P. are employees and stockholders of F. Hoffmann-La Roche Ltd. F.M., V.B., N.P., N.F., and S.F. are employees of Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland. L.Y. is a former employee of the Roche Innovation Center, Little Falls, NJ, USA.

Availability of Data and Material

Qualified researchers may request access to individual patient level data through the clinical study data request platform (https://vivli.org/). Further details on Roche's criteria for eligible studies are available here (https://vivli.org/members/ourmembers/). For further details on Roche's Global Policy on the Sharing of Clinical Information and how to request access to related clinical study documents, see here (https://www.roche.com/research_and_development/who_we_are_how_we_work/clinical_trials/our_commitment_to_data_sharing.htm).

Code Availability

Not applicable.

Authors' Contributions

All authors were involved in interpretation of the data, revising the manuscript critically for important intellectual content, approved the final version, and agree to be accountable for the work. Additionally, the authors contributed as follows: S.F. performed the data analysis; V.B. contributed to the conception and planning of the work that led to the manuscript; N.B. drafted the manuscript content.

Ethics Approval

All studies were approved by the relevant ethics committees, and were conducted in accordance with the principles of the Declaration of Helsinki and Good Clinical Practice guidelines.

Consent to Participate

All subjects provided written informed consent prior to enrollment in the clinical studies.

Consent for Publication

Not applicable.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Djebli, N., Buchheit, V., Parrott, N. et al. Physiologically-Based Pharmacokinetic Modelling of Entrectinib Parent and Active Metabolite to Support Regulatory Decision-Making. Eur J Drug Metab Pharmacokinet 46, 779–791 (2021). https://doi.org/10.1007/s13318-021-00714-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13318-021-00714-z

Navigation