Skip to main content
Log in

Evidence-Based Guidelines for Drug Interaction Studies: Model-Informed Time Course of Intestinal and Hepatic CYP3A4 Inhibition by Clarithromycin

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

Drug-drug interaction (DDI) studies are mandated in drug development; however, protocols for evaluating the impact of cytochrome P450 (CYP) inhibition on new molecular entities are currently inconsistent. This study utilised validated physiologically based pharmacokinetic (PBPK) software to define the optimal dose, frequency, and duration of clarithromycin to achieve optimal characterisation of CYP3A4 inhibition in a study population. The Simcyp® Simulator (Version 19.0) was used to simulate clarithromycin-mediated CYP3A4 inhibition in healthy virtual cohorts. Between trial variability in magnitude and time course of CYP3A4 activity was assessed following clarithromycin dosing strategies obtained from the University of Washington Drug Interaction Database. Heterogeneity in CYP3A4 inhibition was evaluated across sex, race, and age. Literature review identified 500 mg twice daily for 5 days as the most common clarithromycin dosing protocol for CYP3A4 inhibition studies. On simulation, clarithromycin 500 mg twice daily resulted in the largest steady-state inhibition of hepatic (percent mean inhibition [95%CI] = 80 [77–83]) and small intestine (94 [94–95]) CYP3A4 activity (as compared to 500 mg once daily, 400 mg once/twice daily, or 250 mg once/twice daily). Additionally, 500 mg twice daily was associated with the shortest time for 90% of individuals to reach 90% of their minimum hepatic (4 days) and small intestine (1 days) CYP3A4 activity. The study presented herein supports that clarithromycin dosing protocol of 500 mg twice daily for 5 days is sufficient to achieve maximal hepatic and small intestine CYP3A4 inhibition. These findings were consistent between sex, race, and age differences.

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

Similar content being viewed by others

References

  1. Wilkinson GR. Drug metabolism and variability among patients in drug response. The New England journal of medicine. 2005;352(21):2211–21.

    Article  CAS  Google Scholar 

  2. Yan Z, Caldwell GW. Metabolism profiling, and cytochrome P450 inhibition & induction in drug discovery. Current topics in medicinal chemistry. 2001;1(5):403–25.

    Article  CAS  Google Scholar 

  3. Waring MJ, Arrowsmith J, Leach AR, Leeson PD, Mandrell S, Owen RM, Pairaudeau G, Pennie WD, Pickett SD, Wang J, Wallace O, Weir A. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nature Reviews Drug Discovery. 2015;14(7):475–86.

    Article  CAS  Google Scholar 

  4. FDA. Clinical drug interaction studies—cytochrome P450 enzyme- and transporter-mediated drug interactions guidance for industry. 2020.

  5. EMA. Guideline on the investigation of drug interactions. 2012.

  6. Paine MF, Hart HL, Ludington SS, Haining RL, Rettie AE, Zeldin DC. The human intestinal cytochrome P450 “pie”. Drug metabolism and disposition: the biological fate of chemicals. 2006;34(5):880–6.

    Article  CAS  Google Scholar 

  7. Drozdzik M, Busch D, Lapczuk J, Muller J, Ostrowski M, Kurzawski M, et al. Protein abundance of clinically relevant drug-metabolizing enzymes in the human liver and intestine: a comparative analysis in paired tissue specimens. Clin Pharmacol Ther. 2018;104(3):515–24.

    Article  CAS  Google Scholar 

  8. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology & therapeutics. 2013;138(1):103–41.

    Article  CAS  Google Scholar 

  9. Lamba JK, Lin YS, Schuetz EG, Thummel KE. Genetic contribution to variable human CYP3A-mediated metabolism. Advanced drug delivery reviews. 2002;54(10):1271–94.

    Article  CAS  Google Scholar 

  10. Rodrigues D, Rowland A. From endogenous compounds as biomarkers to plasma-derived nanovesicles as liquid biopsy; has the golden age of translational pharmacokinetics-absorption, distribution, metabolism, excretion-drug-drug interaction science finally arrived? Clinical Pharmacology & Therapeutics. 2019;105(6):1407–20.

    Article  CAS  Google Scholar 

  11. Rowland A, Ruanglertboon W, van Dyk M, Wijayakumara D, Wood L, Meech R, et al. Plasma extracellular nanovesicle (exosome) derived biomarkers for ADME pathways: a novel approach to characterise variability in drug exposure. Br J Clin Pharmacol. 2019;85(1):216–26.

    Article  CAS  Google Scholar 

  12. Rodrigues A, van Dyk M, Sorich M, Fahmy A, Useckaite Z, Newman L, et al. Serum-derived extracellular vesicles as liquid biopsy to study the induction of hepatic cytochromes P450 and organic anion transporting polypeptides. Clin Pharmacol Ther. 2021;110:248–58.

    Article  CAS  Google Scholar 

  13. van Dyk M, Kapetas AJ, Hopkins AM, Rodrigues AD, Vourvahis M, Sorich MJ, et al. Validation of a 3-h sampling interval to assess variability in cytochrome P450 3A phenotype and the impact of induction and mechanism-based inhibition using midazolam as a probe substrate. Frontiers in Pharmacology. 2019;10(1120).

  14. Zhou S, Chan SY, Goh BC, Chan E, Duan W, Huang M, et al. Mechanism-based inhibition of cytochrome P450 3A4 by therapeutic drugs. Clinical Pharmacokinetics. 2005;44(3):279–304.

    Article  CAS  Google Scholar 

  15. Kapetas AJ, Sorich MJ, Rodrigues AD, Rowland A. Guidance for rifampin and midazolam dosing protocols to study intestinal and hepatic cytochrome P450 (CYP) 3A4 induction and de-induction. The AAPS journal. 2019;21(5):78.

    Article  Google Scholar 

  16. Rowland Yeo K, Jamei M, Yang J, Tucker GT, Rostami-Hodjegan A. 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 

  17. Badaoui S, Hopkins AM, Rodrigues AD, Miners JO, Sorich MJ, Rowland A. Application of model informed precision dosing to address the impact of pregnancy stage and CYP2D6 phenotype on foetal morphine exposure. The AAPS Journal. 2021;23(1):15.

    Article  CAS  Google Scholar 

  18. Hopkins AM, Wiese MD, Proudman SM, O’Doherty CE, Foster D, Upton RN. Semiphysiologically based pharmacokinetic model of leflunomide disposition in rheumatoid arthritis patients. CPT Pharmacometrics Syst Pharmacol. 2015;4(6):362–71.

    Article  CAS  Google Scholar 

  19. Rowland A, van Dyk M, Hopkins AM, Mounzer R, Polasek TM, Rostami-Hodjegan A, Sorich MJ. Physiologically based pharmacokinetic modeling to identify physiological and molecular characteristics driving variability in drug exposure. Clinical pharmacology and therapeutics. 2018;104(6):1219–28.

    Article  CAS  Google Scholar 

  20. Sorich MJ, Mutlib F, van Dyk M, Hopkins AM, Polasek TM, Marshall JC, et al. Use of physiologically based pharmacokinetic modeling to identify physiological and molecular characteristics driving variability in axitinib exposure: a fresh approach to precision dosing in oncology. Journal of clinical pharmacology. 2019;59(6):872–9.

    Article  CAS  Google Scholar 

  21. Quinney SK, Malireddy SR, Vuppalanchi R, Hamman MA, Chalasani N, Gorski JC, Hall SD. Rate of onset of inhibition of gut-wall and hepatic CYP3A by clarithromycin. European journal of clinical pharmacology. 2013;69(3):439–48.

    Article  CAS  Google Scholar 

  22. Hopkins AM, Rowland A, Sorich MJ. Data sharing from pharmaceutical industry sponsored clinical studies: audit of data availability. BMC medicine. 2018;16(1):165.

    Article  Google Scholar 

  23. Quinney SK, Zhang X, Lucksiri A, Gorski JC, Li L, Hall SD. Physiologically based pharmacokinetic model of mechanism-based inhibition of CYP3A by clarithromycin. Drug Metabolism and Disposition. 2010;38(2):241–8.

    Article  CAS  Google Scholar 

  24. Goldstein EJC, Owens RC Jr, Nolin TD. Antimicrobial-associated QT interval prolongation: pointes of interest. Clinical Infectious Diseases. 2006;43(12):1603–11.

    Article  Google Scholar 

  25. Dresser GK, Spence JD, Bailey DG. Pharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clin pharmacokinet. 2000;38(1):41–57.

    Article  CAS  Google Scholar 

  26. Owens RC Jr. QT prolongation with antimicrobial agents: understanding the significance. Drugs. 2004;64(10):1091–124.

    Article  CAS  Google Scholar 

  27. van Haarst AD, van’t Klooster GA, van Gerven JM, Schoemaker RC, van Oene JC, Burggraaf J, et al. The influence of cisapride and clarithromycin on QT intervals in healthy volunteers. Clin Pharmacol Ther. 1998;64(5):542–6.

    Article  Google Scholar 

  28. Darwich AS, Aslam U, Ashcroft DM, Rostami-Hodjegan A. Meta-analysis of the turnover of intestinal epithelia in preclinical animal species and humans. Drug metabolism and disposition: the biological fate of chemicals. 2014;42(12):2016–22.

    Article  Google Scholar 

  29. Tornio A, Filppula AM, Niemi M, Backman JT. Clinical studies on drug-drug interactions involving metabolism and transport: methodology, pitfalls, and interpretation. Clinical pharmacology and therapeutics. 2019;105(6):1345–61.

    Article  Google Scholar 

  30. Prieto Garcia L, Janzén D, Kanebratt KP, Ericsson H, Lennernäs H, Lundahl A. Physiologically based pharmacokinetic model of itraconazole and two of its metabolites to improve the predictions and the mechanistic understanding of CYP3A4 drug-drug interactions. Drug metabolism and disposition: the biological fate of chemicals. 2018;46(10):1420–33.

    Article  Google Scholar 

  31. Rowland Yeo K, Walsky RL, Jamei M, Rostami-Hodjegan A, Tucker GT. Prediction of time-dependent CYP3A4 drug-drug interactions by physiologically based pharmacokinetic modelling: impact of inactivation parameters and enzyme turnover. Eur J Pharm Sci. 2011;43(3):160–73.

    Article  CAS  Google Scholar 

  32. Rowland A, Mangoni AA, Hopkins A, Sorich MJ, Rowland A. Optimized cocktail phenotyping study protocol using physiological based pharmacokinetic modeling. Front Pharmacol. 2016;7:517.

    Article  Google Scholar 

  33. Rowland A, van Dyk M, Warncken D, Mangoni AA, Sorich MJ, Rowland A. Evaluation of modafinil as a perpetrator of metabolic drug-drug interactions using a model informed cocktail reaction phenotyping trial protocol. Br J Clin Pharmacol. 2017:10.1111/bcp.13478.

  34. Johnson JA. Predictability of the effects of race or ethnicity on pharmacokinetics of drugs. International journal of clinical pharmacology and therapeutics. 2000;38(2):53–60.

    Article  CAS  Google Scholar 

  35. Hunt CM, Westerkam WR, Stave GM. Effect of age and gender on the activity of human hepatic CYP3A. Biochemical pharmacology. 1992;44(2):275–83.

    Article  CAS  Google Scholar 

  36. Venkatakrishnan K, Rostami-Hodjegan A. Come dance with me: transformative changes in the science and practice of drug-drug interactions. Clinical pharmacology and therapeutics. 2019;105(6):1272–8.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ahmad Y Abuhelwa.

Ethics declarations

Competing Interests

A.D.R. is an employee and stockholder of Pfizer Inc. A.R. and M.J.S. are recipients of unrelated research support from Pfizer Inc. All authors declare that there are no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

ESM 1

(DOCX 386 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kapetas, A.J., Abuhelwa, A.Y., Sorich, M.J. et al. Evidence-Based Guidelines for Drug Interaction Studies: Model-Informed Time Course of Intestinal and Hepatic CYP3A4 Inhibition by Clarithromycin. AAPS J 23, 104 (2021). https://doi.org/10.1208/s12248-021-00632-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-021-00632-7

KEY WORDS

Navigation