Abstract

BackgroundDrug-drug interactions resulting from the inhibition of an enzymatic process can have serious implications for clinical drug therapy. Quantification of the drugs internal exposure increase upon administration with an inhibitor requires understanding to avoid the drug reaching toxic thresholds. In this study, we aim to predict the effect of the CYP3A4 inhibitors, itraconazole (ITZ) and its primary metabolite, hydroxyitraconazole (OH-ITZ) on the pharmacokinetics of the anesthetic, midazolam (MDZ) and its metabolites, 1' hydroxymidazolam (1OH-MDZ) and 1' hydroxymidazolam glucuronide (1OH-MDZ-Glu) using mechanistic whole body physiologically-based pharmacokinetic simulation models. The model is build on MDZ, 1OH-MDZ and 1OH-MDZ-Glu plasma concentration time data experimentally determined in 19 CYP3A5 genotyped adult male individuals, who received MDZ intravenously in a basal state. The model is then used to predict MDZ, 1OH-MDZ and 1OH-MDZ-Glu concentrations in an CYP3A-inhibited state following ITZ administration.ResultsFor the basal state model, three linked WB-PBPK models (MDZ, 1OH-MDZ, 1OH-MDZ-Glu) for each individual were elimination optimized that resulted in MDZ and metabolite plasma concentration time curves that matched individual observed clinical data. In vivo Km and Vmax optimized values for MDZ hydroxylation were similar to literature based in vitro measures. With the addition of the ITZ/OH-ITZ model to each individual coupled MDZ + metabolite model, the plasma concentration time curves were predicted to greatly increase the exposure of MDZ as well as to both increase exposure and significantly alter the plasma concentration time curves of the MDZ metabolites in comparison to the basal state curves. As compared to the observed clinical data, the inhibited state curves were generally well described although the simulated concentrations tended to exceed the experimental data between approximately 6 to 12 hours following MDZ administration. This deviations appeared to be greater in the CYP3A5 *1/*1 and CYP3A5 *1/*3 group than in the CYP3A5 *3/*3 group and was potentially the result of assuming that ITZ/OH-ITZ inhibits both CYP3A4 and CYP3A5, whereas in vitro inhibition is due to CYP3A4.ConclusionThis study represents the first attempt to dynamically simulate metabolic enzymatic drug-drug interactions via coupled WB-PBPK models. The workflow described herein, basal state optimization followed by inhibition prediction, is novel and will provide a basis for the development of other inhibitor models that can be used to guide, interpret, and potentially replace clinical drug-drug interaction trials.

Highlights

  • Drug-drug interactions resulting from the inhibition of an enzymatic process can have serious implications for clinical drug therapy

  • The approach that is taken is one which dynamically links inhibitor and drug models using whole-body physiologically-based pharmacokinetic models (WB-PBPK) to quantify, under any administration time and dose regimen, the changes that occur in parent compound exposure as well as the dynamic changes in the respective metabolite exposures

  • This has been done for midazolam (MDZ), and its two major metabolites 1' hydroxymidazolam (1OHMDZ) and the glucuronide of 1' hydroxymidazolam (1OH-MDZ-Glu), in the presence of the CYP3A4 inhibitors itraconazole (ITZ) and its major metabolite hydroxyitraconazole (OH-ITZ)

Read more

Summary

Introduction

Drug-drug interactions resulting from the inhibition of an enzymatic process can have serious implications for clinical drug therapy. The approach that is taken is one which dynamically links inhibitor and drug models using whole-body physiologically-based pharmacokinetic models (WB-PBPK) to quantify, under any administration time and dose regimen, the changes that occur in parent compound exposure as well as the dynamic changes in the respective metabolite exposures. This has been done for midazolam (MDZ), and its two major metabolites 1' hydroxymidazolam (1OHMDZ) and the glucuronide of 1' hydroxymidazolam (1OH-MDZ-Glu), in the presence of the CYP3A4 inhibitors itraconazole (ITZ) and its major metabolite hydroxyitraconazole (OH-ITZ). This example was used because of the importance of CYP3A4 to drug metabolism and the availability of a full clinical data set for MDZ given in the basal and ITZ/OH-ITZ inhibited state [4]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call