メインコンテンツにスキップ

Prediction of Time-dependent CYP3A4 Drug-drug Interactions by Physiologically-based Pharmacokinetic Modeling: Impact of Inactivation Parameters and Enzyme Turnover

Predicting the magnitude of time-dependent metabolic drug-drug (mDDIs) interactions involving cytochrome P-450 3A4 (CYP3A4) from in vitro data requires accurate knowledge of the inactivation parameters of the inhibitor (KI), kinact) and of the turnover of the enzyme (kdeg) in both the gut and the liver. We have predicted the magnitude of mDDIs observed in 29 in vivo studies involving six CYP3A4 probe substrates and five mechanism based inhibitors of CYP3A4 of variable potency (azithromycin, clarithromycin, diltiazem, erythromycin and verapamil). Inactivation parameters determined anew in a single laboratory under standardised conditions together with data from substrate and inhibitor files within the Simcyp® Simulator (Version 9.3) were used to determine a value of the hepatic kdeg (0.0193 or 0.0077h-1) most appropriate for the prediction of mDDIs involving time-dependent inhibition of CYP3A4. The higher value resulted in decreased bias (geometric mean fold error – 1.05 versus 1.30) and increased precision (root mean squared error – 1.29 versus 2.30) of predictions of mean ratios of AUC in the absence and presence of inhibitor. Depending on the kdeg value used (0.0193 versus 0.0077h-1) predicted mean ratios of AUC were within 2-fold of the observed values for all (100%) and 27 (93%) of the 29 studies, respectively and within 1.5-fold for 24 (83%) and 17 (59%) of the 29 studies, respectively. Comprehensive PBPK models were applied for accurate assessment of the potential for mDDIs involving time-dependent inhibition of CYP3A4 using a hepatic kdeg value of 0.0193h-1; in conjunction with inactivation parameters determined by the conventional experimental approach.

Author(s): Karen Rowland Yeo, Robert Walsky, Masoud Jamei, Amin Rostami-Hodjegan, Geoffrey Tucker

Year: 2011年6月14日

Powered by Translations.com GlobalLink OneLink Software