Skip to main content

Advertisement

Log in

Drug–drug interaction predictions with PBPK models and optimal multiresponse sampling time designs: application to midazolam and a phase I compound. Part 2: clinical trial results

  • Published:
Journal of Pharmacokinetics and Pharmacodynamics Aims and scope Submit manuscript

Abstract

Purpose To compare results of population PK analyses obtained with a full empirical design (FD) and an optimal sparse design (MD) in a Drug–Drug Interaction (DDI) study aiming to evaluate the potential CYP3A4 inhibitory effect of a drug in development, SX, on a reference substrate, midazolam (MDZ). Secondary aim was to evaluate the interaction of SX on MDZ in the in vivo study. Methods To compare designs, real data were analysed by population PK modelling technique using either FD or MD with NONMEM FOCEI for SX and with NONMEM FOCEI and MONOLIX SAEM for MDZ. When applicable a Wald test was performed to compare model parameter estimates, such as apparent clearance (CL/F), across designs. To conclude on the potential interaction of SX on MDZ PK, a Student paired test was applied to compare the individual PK parameters (i.e. log(AUC) and log(Cmax)) obtained either by a non-compartmental approach (NCA) using FD or from empirical Bayes estimates (EBE) obtained after fitting the model separately on each treatment group using either FD or MD. Results For SX, whatever the design, CL/F was well estimated and no statistical differences were found between CL/F estimated values obtained with FD (CL/F = 8.2 l/h) and MD (CL/F = 8.2 l/h). For MDZ, only MONOLIX was able to estimate CL/F and to provide its standard error of estimation with MD. With MONOLIX, whatever the design and the administration setting, MDZ CL/F was well estimated and there were no statistical differences between CL/F estimated values obtained with FD (72 l/h and 40 l/h for MDZ alone and for MDZ with SX, respectively) and MD (77 l/h and 45 l/h for MDZ alone and for MDZ with SX, respectively). Whatever the approach, NCA or population PK modelling, and for the latter approach, whatever the design, MD or FD, comparison tests showed that there was a statistical difference (P < 0.0001) between individual MDZ log(AUC) obtained after MDZ administration alone and co-administered with SX. Regarding Cmax, there was a statistical difference (P < 0.05) between individual MDZ log(Cmax) obtained under the 2 administration settings in all cases, except with the sparse design with MONOLIX. However, the effect on Cmax was small. Finally, SX was shown to be a moderate CYP3A4 inhibitor, which at therapeutic doses increased MDZ exposure by a factor of 2 in average and almost did not affect the Cmax. Conclusion The optimal sparse design enabled the estimation of CL/F of a CYP3A4 substrate and inhibitor when co-administered together and to show the interaction leading to the same conclusion as the full empirical design.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Krayenbühl JC, Vozeh S, Kondo-Oestreicher M, Dayer P (1999) Drug–drug interactions of new active substances: mibefradil example. Eur J Clin Pharmacol 55(8):559–565 Review

    Article  PubMed  Google Scholar 

  2. Shah RR (2004) Pharmacogenetic aspects of drug-induced torsade de pointes: potential tool for improving clinical drug development and prescribing. Drug Saf 27(3):145–172. doi:10.2165/00002018-200427030-00001

    Article  PubMed  CAS  Google Scholar 

  3. FDA (2006) Guidance for industry—drug interaction studies—study design, data analysis, and implications for dosing and labeling. http://www.fda.gov/cder/Guidance/6695dft.pdf

  4. Chenel M, Bouzom F, Aarons L, Ogungbenro K (2009) Drug–drug interaction predictions with PBPK models and optimal multiresponse sampling time designs: application to midazolam and a phase I compound. Part 1: comparison of uniresponse and multiresponse designs using PopDes. J Pharmacokinet Pharmacodyn (submitted)

  5. Beal SL, Sheiner LB (1996) NONMEM users guides, version 5, NONMEM project group. University of California, San Francisco

    Google Scholar 

  6. Lavielle M, Mentré F (2007) Estimation of population pharmacokinetic parameters of saquinavir in HIV patients and covariate analysis with the saem algorithm implemented in MONOLIX. J Pharmacokinet Pharmacodyn 34(2):229–249. doi:10.1007/s10928-006-9043-z

    Article  PubMed  CAS  Google Scholar 

  7. Delyon B, Lavielle M, Moulines E (1999) Convergence of a stochastic approximation version of the EM algorithm. Ann Stat 27(1):94–128. doi:10.1214/aos/1018031103

    Article  Google Scholar 

  8. Donnet S, Samson A (2007) Estimation of parameters in incomplete data models defined by dynamical systems. J Stat Plan Infer 137:2815–2831. doi:10.1016/j.jspi.2006.10.013

    Article  Google Scholar 

  9. Kuhn E, Lavielle M (2004) Coupling a stochastic approximation version of EM with a MCMC procedure. ESAIM P&S 8:115–131

    Article  Google Scholar 

  10. Kuhn E, Lavielle M (2005) Maximum likelihood estimation in nonlinear mixed effects models. Comput Stat Data Anal 49(4):1020–1038. doi:10.1016/j.csda.2004.07.002

    Article  Google Scholar 

  11. Comets E, Verstuyft C, Lavielle M, Jaillon P, Becquemont P, Mentré F (2007) Modelling the influence of MDR1 polymorphism on digoxin pharmacokinetic parameters. Eur J Clin Pharmacol 63(5):437–449. doi:10.1007/s00228-007-0269-5

    Article  PubMed  CAS  Google Scholar 

  12. Brendel K, Comets E, Laffont C, Laveille C, Mentré F (2006) Metrics for external model evaluation with an application to the population pharmacokinetics of Gliclazide. Pharm Res 23(9):2036–2049. doi:10.1007/s11095-006-9067-5

    Article  PubMed  CAS  Google Scholar 

  13. Savic RM, Karlsson MO (2007) Importance of shrinkage in Epirical Bayes estimates for diagnostics and estimation: problems and solutions. Abstr 1087, p. 16

    Google Scholar 

  14. Panhard X, Mentré F (2005) Evaluation by simulation of tests based on non-linear mixed-effects models in pharmacokinetic interaction and bioequivalence cross-over trials. Stat Med 24:1509–1524. doi:10.1002/sim.2047

    Article  PubMed  Google Scholar 

  15. Panhard X, Taburet A-M, Piketti C, Mentré F (2007) Impact of modelling intra-subject variability on tests based on non-linear mixed-effects models in cross-over pharmacokinetic trials with application to the interaction of tenofovir on atazanavir in HIV patients. Stat Med 26:1268–1284. doi:10.1002/sim.2622

    Article  PubMed  Google Scholar 

  16. Gueorguieva I, Ogungbenro K, Graham G, Glatt S, Aarons L (2007) A program for individual and population optimal design for univariate and multivariate response pharmacokinetic and pharmacodynamic models. Comput Methods Programs Biomed 86(1):51–61. doi:10.1016/j.cmpb.2007.01.004

    Article  PubMed  Google Scholar 

  17. FDA (1999) Guidance for industry—population pharmacokinetics. http://www.fda.gov/cder/guidance/1852fnl.pdf

  18. FDA (2003) Guidance for industry—bioavailability and bioequivalence studies for orally administered drug products—general considerations. http://www.fda.gov/cder/guidance/5356fnl.pdf

  19. EMEA (2002) Note for guidance on the investigation of bioavailability and bioequivalence. http://www.emea.europa.eu/pdfs/human/ewp/140198en.pdf

  20. EMEA (1998) Note for guidance on the investigation of drug interactions. http://www.emea.europa.eu/pdfs/human/ewp/056095en.pdf

  21. Björkman S, Wada DR, Berling BM, Benoni G (2001) Prediction of the disposition of midazolam in surgical patients by a physiologically based pharmacokinetic model. J Pharm Sci 90(9):1226–1241. doi:10.1002/jps.1076

    Article  PubMed  Google Scholar 

  22. Tod M, Lagneau F, Jullien V, Mimoz O (2008) A physiological model to evaluate drug kinetics in patients with hemorrhagic shock followed by fluid resuscitation: application to amoxicillin-clavulanate. Pharm Res 25(6):1431–1439

    Google Scholar 

Download references

Acknowledgement

We would like to acknowledge the Centre for Applied Pharmacokinetic Research (the CAPKR is supported by the following consortium members: Eli Lilly, GlaxoSmithKline, Novartis, Pfizer and Servier).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marylore Chenel.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chenel, M., Bouzom, F., Cazade, F. et al. Drug–drug interaction predictions with PBPK models and optimal multiresponse sampling time designs: application to midazolam and a phase I compound. Part 2: clinical trial results. J Pharmacokinet Pharmacodyn 35, 661–681 (2008). https://doi.org/10.1007/s10928-008-9105-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10928-008-9105-5

Keywords

Navigation