Skip to main content
Top
Published in: Clinical Pharmacokinetics 11/2010

01-11-2010 | Current Opinion

A Quantitative Framework and Strategies for Management and Evaluation of Metabolic Drug-Drug Interactions in Oncology Drug Development

New Molecular Entities as Object Drugs

Authors: Dr Karthik Venkatakrishnan, PhD, Michael D. Pickard, Lisa L. von Moltke

Published in: Clinical Pharmacokinetics | Issue 11/2010

Login to get access

Abstract

This article outlines general strategies for the management and evaluation of pharmacokinetic drug-drug interactions (DDIs) resulting from perturbation of clearance of investigational anticancer drug candidates by concomitantly administered agents in a drug development setting, with a focus on drug candidates that cannot be evaluated in first-in-human studies in healthy subjects. A risk level classification is proposed, based on quantitative integration of knowledge derived from preclinical drug-metabolism studies evaluating the projected percentage contribution [fi(%)] of individual molecular determinants (e.g. cytochrome P450 isoenzymes) to the overall human clearance of the investigational agent. The following classification is proposed with respect to susceptibility to DDIs with metabolic inhibitors: a projected maximum DDI expected to result in a ≤1.33-fold increase in exposure, representing a low level of risk; a projected maximum DDI expected to result in a >1.33-fold but <2-fold increase in exposure, representing a moderate level of risk; and a projected maximum DDI expected to result in a ≥2-fold increase in exposure, representing a potentially high level of risk.
For DDIs with metabolic inducers, the following operational classification is proposed, based on the sum of the percentage contributions of enzymes that are inducible via a common mechanism to the overall clearance of the investigational drug: ≪25%, representing a low level of risk; <50%, representing a moderate level of risk; and ≥50%, representing a potentially high level of risk. To ensure patient safety and to minimize bias in determination of the recommended phase II dose (RP2D), it is recommended that strong and moderate inhibitors and inducers of the major contributing enzyme are excluded in phase I dose-escalation studies of high-risk compounds, whereas exclusion of strong inhibitors and inducers of the contributing enzyme (s) is recommended as being sufficient for moderate-risk compounds. For drugs that will be investigated in diseases such as glioblastoma, where there may be relatively frequent use of enzyme-inducing antiepileptic agents (EIAEDs), a separate dose-escalation study in this subpopulation is recommended to define the RP2D. For compounds in the high-risk category, if genetic deficiencies in the activity of the major drug-metabolizing enzyme are known, it is recommended that poor metabolizers be studied separately to define the RP2D for this subpopulation.
Whereas concomitant medication exclusion criteria that are utilized in the phase I dose-escalation studies will probably also need to be maintained for high-risk compounds in phase II studies unless the results of a clinical DDI study indicate the absence of a clinically relevant interaction, these exclusion criteria can potentially be relaxed beyond phase I for moderate-risk compounds, if supported by the nature of clinical toxicities and the understanding of the therapeutic index in phase I. Adequately designed clinical DDI studies will not only inform potential relaxation of concomitant medication exclusion criteria in later-phase studies but, importantly, will also inform the development of pharmacokinetically derived dose-modification guidelines for use in clinical practice when coupled with adequate safety monitoring, as illustrated in the prescribing guidance for many recently approved oncology therapeutics.
Literature
1.
go back to reference Huang SM, Lesko LJ. Drug-drug, drug-dietary supplement, and drug-citrus fruit and other food interactions: what have we learned? J Clin Pharmacol 2004; 44(6): 559–69PubMedCrossRef Huang SM, Lesko LJ. Drug-drug, drug-dietary supplement, and drug-citrus fruit and other food interactions: what have we learned? J Clin Pharmacol 2004; 44(6): 559–69PubMedCrossRef
2.
go back to reference Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct of in vitro and in vivo drug-drug interaction studies: a Pharmaceutical Research and Manufacturers of America (PhRMA) perspective. Drug Metab Dispos 2003; 31(7): 815–32PubMedCrossRef Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct of in vitro and in vivo drug-drug interaction studies: a Pharmaceutical Research and Manufacturers of America (PhRMA) perspective. Drug Metab Dispos 2003; 31(7): 815–32PubMedCrossRef
3.
go back to reference Huang SM, Strong JM, Zhang L, et al. New era in drug interaction evaluation: US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. J Clin Pharmacol 2008; 48(6): 662–70PubMedCrossRef Huang SM, Strong JM, Zhang L, et al. New era in drug interaction evaluation: US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. J Clin Pharmacol 2008; 48(6): 662–70PubMedCrossRef
4.
go back to reference Huang SM, Temple R, Throckmorton DC, et al. Drug interaction studies: study design, data analysis, and implications for dosing and labeling. Clin Pharmacol Ther 2007; 81(2): 298–304PubMedCrossRef Huang SM, Temple R, Throckmorton DC, et al. Drug interaction studies: study design, data analysis, and implications for dosing and labeling. Clin Pharmacol Ther 2007; 81(2): 298–304PubMedCrossRef
5.
go back to reference Tucker GT, Houston JB, Huang SM. Optimizing drug development: strategies to assess drug metabolism/transporter interaction potential — towards a consensus. Br J Clin Pharmacol 2001; 52(1): 107–17PubMedCrossRef Tucker GT, Houston JB, Huang SM. Optimizing drug development: strategies to assess drug metabolism/transporter interaction potential — towards a consensus. Br J Clin Pharmacol 2001; 52(1): 107–17PubMedCrossRef
6.
go back to reference Zhang L, Reynolds KS, Zhao P, et al. Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 2010; 243(2): 134–45PubMedCrossRef Zhang L, Reynolds KS, Zhao P, et al. Drug interactions evaluation: an integrated part of risk assessment of therapeutics. Toxicol Appl Pharmacol 2010; 243(2): 134–45PubMedCrossRef
15.
go back to reference Dutreix C, Peng B, Mehring G, et al. Pharmacokinetic interaction between ketoconazole and imatinib mesylate (Glivec) in healthy subjects. Cancer Chemother Pharmacol 2004; 54(4): 290–4PubMedCrossRef Dutreix C, Peng B, Mehring G, et al. Pharmacokinetic interaction between ketoconazole and imatinib mesylate (Glivec) in healthy subjects. Cancer Chemother Pharmacol 2004; 54(4): 290–4PubMedCrossRef
17.
go back to reference Boni JP, Leister C, Burns J, et al. Differential effects of ketoconazole on exposure to temsirolimus following intravenous infusion of temsirolimus. Br J Cancer 2008; 98(11): 1797–802PubMedCrossRef Boni JP, Leister C, Burns J, et al. Differential effects of ketoconazole on exposure to temsirolimus following intravenous infusion of temsirolimus. Br J Cancer 2008; 98(11): 1797–802PubMedCrossRef
18.
go back to reference Van Erp NP, Gelderblom H, Karlsson MO, et al. Influence of CYP3A4 inhibition on the steady-state pharmacokinetics of imatinib. Clin Cancer Res 2007; 13(24): 7394–400PubMedCrossRef Van Erp NP, Gelderblom H, Karlsson MO, et al. Influence of CYP3A4 inhibition on the steady-state pharmacokinetics of imatinib. Clin Cancer Res 2007; 13(24): 7394–400PubMedCrossRef
19.
go back to reference O’Brien SG, Meinhardt P, Bond E, et al. Effects of imatinib mesylate (STI571, Glivec) on the pharmacokinetics of simvastatin, a cytochrome p450 3A4 substrate, in patients with chronic myeloid leukaemia. Br J Cancer 2003; 89(10): 1855–9PubMedCrossRef O’Brien SG, Meinhardt P, Bond E, et al. Effects of imatinib mesylate (STI571, Glivec) on the pharmacokinetics of simvastatin, a cytochrome p450 3A4 substrate, in patients with chronic myeloid leukaemia. Br J Cancer 2003; 89(10): 1855–9PubMedCrossRef
20.
go back to reference Venkatakrishnan K, Rader M, Ramanathan RK, et al. Effect of the CYP3A inhibitor ketoconazole on the pharmacokinetics and pharmacodynamics of bortezomib in patients with advanced solid tumors: a prospective, multicenter, open-label, randomized, two-way crossover drug-drug interaction study. Clin Ther 2009; 31(Pt 2): 2444–58PubMedCrossRef Venkatakrishnan K, Rader M, Ramanathan RK, et al. Effect of the CYP3A inhibitor ketoconazole on the pharmacokinetics and pharmacodynamics of bortezomib in patients with advanced solid tumors: a prospective, multicenter, open-label, randomized, two-way crossover drug-drug interaction study. Clin Ther 2009; 31(Pt 2): 2444–58PubMedCrossRef
21.
go back to reference Engels FK, Ten Tije AJ, Baker SD, et al. Effect of cytochrome P450 3A4 inhibition on the pharmacokinetics of docetaxel. Clin Pharmacol Ther 2004; 75(5): 448–54PubMedCrossRef Engels FK, Ten Tije AJ, Baker SD, et al. Effect of cytochrome P450 3A4 inhibition on the pharmacokinetics of docetaxel. Clin Pharmacol Ther 2004; 75(5): 448–54PubMedCrossRef
22.
go back to reference Johnson FM, Agrawal S, Burris H, et al. Phase 1 pharmacokinetic and druginteraction study of dasatinib in patients with advanced solid tumors. Cancer 2010; 116(6): 1582–91PubMedCrossRef Johnson FM, Agrawal S, Burris H, et al. Phase 1 pharmacokinetic and druginteraction study of dasatinib in patients with advanced solid tumors. Cancer 2010; 116(6): 1582–91PubMedCrossRef
23.
go back to reference Goel S, Cohen M, Comezoglu SN, et al. The effect of ketoconazole on the pharmacokinetics and pharmacodynamics of ixabepilone: a first in class epothilone B analogue in late-phase clinical development. Clin Cancer Res 2008; 14(9): 2701–9PubMedCrossRef Goel S, Cohen M, Comezoglu SN, et al. The effect of ketoconazole on the pharmacokinetics and pharmacodynamics of ixabepilone: a first in class epothilone B analogue in late-phase clinical development. Clin Cancer Res 2008; 14(9): 2701–9PubMedCrossRef
24.
go back to reference Hisaka A, Kusama M, Ohno Y, et al. A proposal for a pharmacokinetic interaction significance classification system (PISCS) based on predicted drug exposure changes and its potential application to alert classifications in product labelling. Clin Pharmacokinet 2009; 48(10): 653–66PubMedCrossRef Hisaka A, Kusama M, Ohno Y, et al. A proposal for a pharmacokinetic interaction significance classification system (PISCS) based on predicted drug exposure changes and its potential application to alert classifications in product labelling. Clin Pharmacokinet 2009; 48(10): 653–66PubMedCrossRef
25.
go back to reference Ito K, Hallifax D, Obach RS, et al. Impact of parallel pathways of drug elimination and multiple cytochrome P450 involvement on drug-drug interactions: CYP2D6 paradigm. Drug Metab Dispos 2005; 33(6): 837–44PubMedCrossRef Ito K, Hallifax D, Obach RS, et al. Impact of parallel pathways of drug elimination and multiple cytochrome P450 involvement on drug-drug interactions: CYP2D6 paradigm. Drug Metab Dispos 2005; 33(6): 837–44PubMedCrossRef
26.
go back to reference Lu C, Hatsis P, Berg C, et al. Prediction of pharmacokinetic drug-drug interactions using human hepatocyte suspension in plasma and cytochrome P450 phenotypic data: II. In vitro-in vivo correlation with ketoconazole. Drug Metab Dispos 2008; 36(7): 1255–60PubMedCrossRef Lu C, Hatsis P, Berg C, et al. Prediction of pharmacokinetic drug-drug interactions using human hepatocyte suspension in plasma and cytochrome P450 phenotypic data: II. In vitro-in vivo correlation with ketoconazole. Drug Metab Dispos 2008; 36(7): 1255–60PubMedCrossRef
27.
go back to reference Lu C, Miwa GT, Prakash SR, et al. A novel model for the prediction of drugdrug interactions in humans based on in vitro cytochrome P450 phenotypic data. Drug Metab Dispos 2007; 35(1): 79–85PubMedCrossRef Lu C, Miwa GT, Prakash SR, et al. A novel model for the prediction of drugdrug interactions in humans based on in vitro cytochrome P450 phenotypic data. Drug Metab Dispos 2007; 35(1): 79–85PubMedCrossRef
28.
go back to reference Obach RS, Walsky RL, Venkatakrishnan K, et al. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug-drug interactions. J Pharmacol Exp Ther 2006; 316(1): 336–48PubMedCrossRef Obach RS, Walsky RL, Venkatakrishnan K, et al. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug-drug interactions. J Pharmacol Exp Ther 2006; 316(1): 336–48PubMedCrossRef
29.
go back to reference Obach RS, Walsky RL, Venkatakrishnan K, et al. In vitro cytochrome P450 inhibition data and the prediction of drug-drug interactions: qualitative relationships, quantitative predictions, and the rank-order approach. Clin Pharmacol Ther 2005; 78(6): 582–92PubMedCrossRef Obach RS, Walsky RL, Venkatakrishnan K, et al. In vitro cytochrome P450 inhibition data and the prediction of drug-drug interactions: qualitative relationships, quantitative predictions, and the rank-order approach. Clin Pharmacol Ther 2005; 78(6): 582–92PubMedCrossRef
30.
go back to reference Ohno Y, Hisaka A, Suzuki H. General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase bycoadministrationofstandarddrugs. Clin Pharmacokinet 2007; 46(8): 681–96PubMedCrossRef Ohno Y, Hisaka A, Suzuki H. General framework for the quantitative prediction of CYP3A4-mediated oral drug interactions based on the AUC increase bycoadministrationofstandarddrugs. Clin Pharmacokinet 2007; 46(8): 681–96PubMedCrossRef
31.
go back to reference Ohno Y, Hisaka A, Ueno M, et al. General framework for the predictionoforal drug interactions caused by CYP3A4 induction from in vivo information. Clin Pharmacokinet 2008; 47(10): 669–80PubMedCrossRef Ohno Y, Hisaka A, Ueno M, et al. General framework for the predictionoforal drug interactions caused by CYP3A4 induction from in vivo information. Clin Pharmacokinet 2008; 47(10): 669–80PubMedCrossRef
32.
go back to reference Youdim KA, Zayed A, Dickins M, et al. Application of CYP3A4 in vitro data to predict clinical drug-drug interactions; predictionsofcompounds asobjects of interaction. Br J Clin Pharmacol 2008; 65(5): 680–92PubMedCrossRef Youdim KA, Zayed A, Dickins M, et al. Application of CYP3A4 in vitro data to predict clinical drug-drug interactions; predictionsofcompounds asobjects of interaction. Br J Clin Pharmacol 2008; 65(5): 680–92PubMedCrossRef
33.
go back to reference Zhang H, Davis CD, Sinz MW, et al. Cytochrome P450 reaction-phenotyping: an industrial perspective. Expert Opin Drug Metab Toxicol 2007; 3(5): 667–87PubMedCrossRef Zhang H, Davis CD, Sinz MW, et al. Cytochrome P450 reaction-phenotyping: an industrial perspective. Expert Opin Drug Metab Toxicol 2007; 3(5): 667–87PubMedCrossRef
34.
go back to reference Miners JO, Mackenzie PI, Knights KM. The prediction of drug-glucuronidation parameters in humans: UDP-glucuronosyltransferase enzyme-selective substrate and inhibitor probes for reaction phenotyping and in vitro-in vivo extrapolation of drug clearance and drug-drug interaction potential. Drug Metab Rev 2010; 42(1): 189–201CrossRef Miners JO, Mackenzie PI, Knights KM. The prediction of drug-glucuronidation parameters in humans: UDP-glucuronosyltransferase enzyme-selective substrate and inhibitor probes for reaction phenotyping and in vitro-in vivo extrapolation of drug clearance and drug-drug interaction potential. Drug Metab Rev 2010; 42(1): 189–201CrossRef
35.
go back to reference Kilford PJ, Stringer R, Sohal B, et al. Prediction of drug clearance by glucuronidation from in vitro data: use of combined cytochrome P450 and UDP-glucuronosyltransferase cofactors in alamethicin-activated human liver microsomes. Drug Metab Dispos 2009; 37(1): 82–9PubMedCrossRef Kilford PJ, Stringer R, Sohal B, et al. Prediction of drug clearance by glucuronidation from in vitro data: use of combined cytochrome P450 and UDP-glucuronosyltransferase cofactors in alamethicin-activated human liver microsomes. Drug Metab Dispos 2009; 37(1): 82–9PubMedCrossRef
36.
go back to reference Balliet RM, Chen G, Gallagher CJ, et al. Characterization of UGTs active against SAHA and association between SAHA glucuronidation activity phenotype with UGT genotype. Cancer Res 2009; 69(7): 2981–9PubMedCrossRef Balliet RM, Chen G, Gallagher CJ, et al. Characterization of UGTs active against SAHA and association between SAHA glucuronidation activity phenotype with UGT genotype. Cancer Res 2009; 69(7): 2981–9PubMedCrossRef
37.
go back to reference Iyer L, Hall D, Das S, et al. Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther 1999; 65(5): 576–82PubMedCrossRef Iyer L, Hall D, Das S, et al. Phenotype-genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin Pharmacol Ther 1999; 65(5): 576–82PubMedCrossRef
38.
go back to reference Peterkin VC, Bauman JN, Goosen TC, et al. Limited influence of UGT1A1*28 and no effect of UGT2B7*2 polymorphisms on UGT1A1 or UGT2B7 activities and protein expression in human liver microsomes. Br J Clin Pharmacol 2007; 64(4): 458–68PubMedCrossRef Peterkin VC, Bauman JN, Goosen TC, et al. Limited influence of UGT1A1*28 and no effect of UGT2B7*2 polymorphisms on UGT1A1 or UGT2B7 activities and protein expression in human liver microsomes. Br J Clin Pharmacol 2007; 64(4): 458–68PubMedCrossRef
39.
go back to reference Kirby BJ, Unadkat JD. The impact of ignoring extraction ratio when predicting drug-drug interactions, fraction metabolized and intestinal first-pass contribution. Drug Metab Dispos. Epub 2010 Aug 19 Kirby BJ, Unadkat JD. The impact of ignoring extraction ratio when predicting drug-drug interactions, fraction metabolized and intestinal first-pass contribution. Drug Metab Dispos. Epub 2010 Aug 19
40.
go back to reference Galetin A, Gertz M, Houston JB. Potential role of intestinal first-pass metabolism in the prediction of drug-drug interactions. Expert Opin Drug Metab Toxicol 2008; 4(7): 909–22PubMedCrossRef Galetin A, Gertz M, Houston JB. Potential role of intestinal first-pass metabolism in the prediction of drug-drug interactions. Expert Opin Drug Metab Toxicol 2008; 4(7): 909–22PubMedCrossRef
41.
go back to reference Galetin A, Hinton LK, Burt H, et al. Maximal inhibition of intestinal first-pass metabolism as a pragmatic indicator of intestinal contribution to the drug-drug interactions for CYP3A4 cleared drugs. Curr Drug Metab 2007; 8(7): 685–93PubMedCrossRef Galetin A, Hinton LK, Burt H, et al. Maximal inhibition of intestinal first-pass metabolism as a pragmatic indicator of intestinal contribution to the drug-drug interactions for CYP3A4 cleared drugs. Curr Drug Metab 2007; 8(7): 685–93PubMedCrossRef
42.
go back to reference Wang YH, Jones DR, Hall SD. Prediction of cytochrome P450 3A inhibition by verapamil enantiomers and their metabolites. Drug Metab Dispos 2004; 32(2): 259–66PubMedCrossRef Wang YH, Jones DR, Hall SD. Prediction of cytochrome P450 3A inhibition by verapamil enantiomers and their metabolites. Drug Metab Dispos 2004; 32(2): 259–66PubMedCrossRef
43.
go back to reference Tang H, Mayersohn M. A global examination of allometric scaling for predicting human drug clearance and the prediction of large vertical allometry. J Pharm Sci 2006; 95(8): 1783–99PubMedCrossRef Tang H, Mayersohn M. A global examination of allometric scaling for predicting human drug clearance and the prediction of large vertical allometry. J Pharm Sci 2006; 95(8): 1783–99PubMedCrossRef
44.
go back to reference Abe K, Bridges AS, Brouwer KL. Use of sandwich-cultured human hepatocytes to predict biliary clearance of angiotensin II receptor blockers and HMG-CoA reductase inhibitors. Drug Metab Dispos 2009; 37(3): 447–52PubMedCrossRef Abe K, Bridges AS, Brouwer KL. Use of sandwich-cultured human hepatocytes to predict biliary clearance of angiotensin II receptor blockers and HMG-CoA reductase inhibitors. Drug Metab Dispos 2009; 37(3): 447–52PubMedCrossRef
45.
go back to reference Ghibellini G, Vasist LS, Leslie EM, et al. In vitro-in vivo correlation of hepatobiliary drug clearance in humans. Clin Pharmacol Ther 2007; 81(3): 406–13PubMedCrossRef Ghibellini G, Vasist LS, Leslie EM, et al. In vitro-in vivo correlation of hepatobiliary drug clearance in humans. Clin Pharmacol Ther 2007; 81(3): 406–13PubMedCrossRef
46.
go back to reference 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. Drug Metab Pharmacokinet 2009; 24(1): 53–75PubMedCrossRef 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. Drug Metab Pharmacokinet 2009; 24(1): 53–75PubMedCrossRef
47.
go back to reference Jamei M, Marciniak S, Feng K, et al. The Simcyp® population-based ADME simulator. Expert Opin Drug Metab Toxicol 2009; 5(2): 211–23PubMedCrossRef Jamei M, Marciniak S, Feng K, et al. The Simcyp® population-based ADME simulator. Expert Opin Drug Metab Toxicol 2009; 5(2): 211–23PubMedCrossRef
48.
go back to reference Rakhit A, Pantze MP, Fettner S, et al. The effects of CYP3A4 inhibition on erlotinib pharmacokinetics: computer-based simulation (Simcyp) predicts in vivo metabolic inhibition. Eur J Clin Pharmacol 2008; 64(1): 31–41PubMedCrossRef Rakhit A, Pantze MP, Fettner S, et al. The effects of CYP3A4 inhibition on erlotinib pharmacokinetics: computer-based simulation (Simcyp) predicts in vivo metabolic inhibition. Eur J Clin Pharmacol 2008; 64(1): 31–41PubMedCrossRef
49.
go back to reference Rostami-Hodjegan A, Tucker G. ’In silico’ simulations to assess the ‘in vivo’ consequences of ‘in vitro’ metabolic drug-drug interactions. Drug Discovery Today: Technologies 2004; 1(4): 441–8CrossRef Rostami-Hodjegan A, Tucker G. ’In silico’ simulations to assess the ‘in vivo’ consequences of ‘in vitro’ metabolic drug-drug interactions. Drug Discovery Today: Technologies 2004; 1(4): 441–8CrossRef
50.
go back to reference Rostami-Hodjegan A, Tucker GT. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 2007; 6(2): 140–8PubMedCrossRef Rostami-Hodjegan A, Tucker GT. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 2007; 6(2): 140–8PubMedCrossRef
51.
go back to reference Roffey SJ, Obach RS, Gedge JI, et al. What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs. Drug Metab Rev 2007; 39(1): 17–43PubMedCrossRef Roffey SJ, Obach RS, Gedge JI, et al. What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs. Drug Metab Rev 2007; 39(1): 17–43PubMedCrossRef
52.
go back to reference Benet LZ. Predicting drug disposition via application of a Biopharmaceutics Drug Disposition Classification System. Basic Clin Pharmacol Toxicol 2010; 106(3): 162–7PubMedCrossRef Benet LZ. Predicting drug disposition via application of a Biopharmaceutics Drug Disposition Classification System. Basic Clin Pharmacol Toxicol 2010; 106(3): 162–7PubMedCrossRef
53.
go back to reference Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010; 9(3): 215–36PubMedCrossRef Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010; 9(3): 215–36PubMedCrossRef
54.
go back to reference Wu CY, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res 2005; 22(1): 11–23PubMedCrossRef Wu CY, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res 2005; 22(1): 11–23PubMedCrossRef
55.
go back to reference Benet LZ. The drug transporter-metabolism alliance: uncovering and defining the interplay. Mol Pharm 2009; 6(6): 1631–43PubMedCrossRef Benet LZ. The drug transporter-metabolism alliance: uncovering and defining the interplay. Mol Pharm 2009; 6(6): 1631–43PubMedCrossRef
56.
go back to reference Hirano M, Maeda K, Shitara Y, et al. Contribution ofOATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther 2004; 311(1): 139–46PubMedCrossRef Hirano M, Maeda K, Shitara Y, et al. Contribution ofOATP2 (OATP1B1) and OATP8 (OATP1B3) to the hepatic uptake of pitavastatin in humans. J Pharmacol Exp Ther 2004; 311(1): 139–46PubMedCrossRef
57.
go back to reference Shitara Y, Sato H, Sugiyama Y. Evaluation of drug-drug interaction in the hepatobiliary and renal transport of drugs. Annu Rev Pharmacol Toxicol 2005; 45: 689–723PubMedCrossRef Shitara Y, Sato H, Sugiyama Y. Evaluation of drug-drug interaction in the hepatobiliary and renal transport of drugs. Annu Rev Pharmacol Toxicol 2005; 45: 689–723PubMedCrossRef
58.
go back to reference Yamada A, Maeda K, Kamiyama E, et al. Multiple human isoforms of drug transporters contribute to the hepatic and renal transport of olmesartan, a selective antagonist of the angiotensin II AT1-receptor. Drug Metab Dispos 2007; 35(12): 2166–76PubMedCrossRef Yamada A, Maeda K, Kamiyama E, et al. Multiple human isoforms of drug transporters contribute to the hepatic and renal transport of olmesartan, a selective antagonist of the angiotensin II AT1-receptor. Drug Metab Dispos 2007; 35(12): 2166–76PubMedCrossRef
59.
go back to reference Yamashiro W, Maeda K, Hirouchi M, et al. Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II AT1-receptor, in humans. Drug Metab Dispos 2006; 34(7): 1247–54PubMedCrossRef Yamashiro W, Maeda K, Hirouchi M, et al. Involvement of transporters in the hepatic uptake and biliary excretion of valsartan, a selective antagonist of the angiotensin II AT1-receptor, in humans. Drug Metab Dispos 2006; 34(7): 1247–54PubMedCrossRef
60.
go back to reference Poirier A, Cascais AC, Funk C, et al. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn 2009; 36(6): 585–611PubMedCrossRef Poirier A, Cascais AC, Funk C, et al. Prediction of pharmacokinetic profile of valsartan in human based on in vitro uptake transport data. J Pharmacokinet Pharmacodyn 2009; 36(6): 585–611PubMedCrossRef
61.
go back to reference Poirier A, Funk C, Scherrmann JM, et al. Mechanistic modeling of hepatic transport from cells to whole body: application to napsagatran and fexofenadine. Mol Pharm 2009; 6(6): 1716–33PubMedCrossRef Poirier A, Funk C, Scherrmann JM, et al. Mechanistic modeling of hepatic transport from cells to whole body: application to napsagatran and fexofenadine. Mol Pharm 2009; 6(6): 1716–33PubMedCrossRef
62.
go back to reference Watanabe T, Kusuhara H, Maeda K, et al. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. J Pharmacol Exp Ther 2009; 328(2): 652–62PubMedCrossRef Watanabe T, Kusuhara H, Maeda K, et al. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. J Pharmacol Exp Ther 2009; 328(2): 652–62PubMedCrossRef
63.
go back to reference Urquhart BL, Tirona RG, Kim RB. Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interin-dividual variability in response to drugs. J Clin Pharmacol 2007; 47(5): 566–78PubMedCrossRef Urquhart BL, Tirona RG, Kim RB. Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interin-dividual variability in response to drugs. J Clin Pharmacol 2007; 47(5): 566–78PubMedCrossRef
64.
go back to reference Hewitt NJ, Lecluyse EL, Ferguson SS. Induction of hepatic cytochrome P450 enzymes: methods, mechanisms, recommendations, and in vitro-in vivo correlations. Xenobiotica 2007; 37(10–11): 1196–224PubMedCrossRef Hewitt NJ, Lecluyse EL, Ferguson SS. Induction of hepatic cytochrome P450 enzymes: methods, mechanisms, recommendations, and in vitro-in vivo correlations. Xenobiotica 2007; 37(10–11): 1196–224PubMedCrossRef
65.
go back to reference Rae JM, Johnson MD, Lippman ME, et al. Rifampin is a selective, pleiotropic inducer of drug metabolism genes in human hepatocytes: studies with cDNA and oligonucleotide expression arrays. J Pharmacol Exp Ther 2001; 299(3): 849–57PubMed Rae JM, Johnson MD, Lippman ME, et al. Rifampin is a selective, pleiotropic inducer of drug metabolism genes in human hepatocytes: studies with cDNA and oligonucleotide expression arrays. J Pharmacol Exp Ther 2001; 299(3): 849–57PubMed
66.
go back to reference Gandelman K, Zhu T, Fahmi OA, et al. Unexpected effect of rifampin on the pharmacokinetics of linezolid: in silico and in vitro approaches to explain its mechanism. J Clin Pharmacol. Epub 2010 Apr 6 Gandelman K, Zhu T, Fahmi OA, et al. Unexpected effect of rifampin on the pharmacokinetics of linezolid: in silico and in vitro approaches to explain its mechanism. J Clin Pharmacol. Epub 2010 Apr 6
67.
go back to reference Yang J, Jamei M, Yeo KR, et al. Prediction of intestinal first-pass drug metabolism. Curr Drug Metab 2007; 8(7): 676–84PubMedCrossRef Yang J, Jamei M, Yeo KR, et al. Prediction of intestinal first-pass drug metabolism. Curr Drug Metab 2007; 8(7): 676–84PubMedCrossRef
68.
go back to reference Almond LM, Yang J, Jamei M, et al. Towards aquantitative framework for the prediction of DDIs arising from cytochrome P450 induction. Curr Drug Metab 2009; 10(4): 420–32PubMedCrossRef Almond LM, Yang J, Jamei M, et al. Towards aquantitative framework for the prediction of DDIs arising from cytochrome P450 induction. Curr Drug Metab 2009; 10(4): 420–32PubMedCrossRef
69.
go back to reference Chu V, Einolf HJ, Evers R, et al. In vitro and in vivo induction of cytochrome P450: a survey of the current practices and recommendations: a Pharmaceutical Research and Manufacturers of America Perspective. Drug Metab Dispos 2009; 37(7): 1339–54PubMedCrossRef Chu V, Einolf HJ, Evers R, et al. In vitro and in vivo induction of cytochrome P450: a survey of the current practices and recommendations: a Pharmaceutical Research and Manufacturers of America Perspective. Drug Metab Dispos 2009; 37(7): 1339–54PubMedCrossRef
70.
go back to reference Mahr G, Sorgel F, Granneman GR, et al. Effects of temafloxacin and ciprofloxacin on the pharmacokinetics of caffeine. Clin Pharmacokinet 1992; 22 Suppl. 1: 90–7PubMedCrossRef Mahr G, Sorgel F, Granneman GR, et al. Effects of temafloxacin and ciprofloxacin on the pharmacokinetics of caffeine. Clin Pharmacokinet 1992; 22 Suppl. 1: 90–7PubMedCrossRef
71.
go back to reference Granfors MT, Backman JT, Neuvonen M, et al. Ciprofloxacin greatly increases concentrations and hypotensive effect of tizanidine by inhibiting its cytochrome P450 1A2-mediated presystemic metabolism. Clin Pharmacol Ther 2004; 76(6): 598–606PubMedCrossRef Granfors MT, Backman JT, Neuvonen M, et al. Ciprofloxacin greatly increases concentrations and hypotensive effect of tizanidine by inhibiting its cytochrome P450 1A2-mediated presystemic metabolism. Clin Pharmacol Ther 2004; 76(6): 598–606PubMedCrossRef
72.
go back to reference Turpeinen M, Tolonen A, Uusitalo J, et al. Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Clin Pharmacol Ther 2005; 77(6): 553–9PubMedCrossRef Turpeinen M, Tolonen A, Uusitalo J, et al. Effect of clopidogrel and ticlopidine on cytochrome P450 2B6 activity as measured by bupropion hydroxylation. Clin Pharmacol Ther 2005; 77(6): 553–9PubMedCrossRef
73.
go back to reference Preskorn SH. Clinically relevant pharmacology of selective serotonin reuptake inhibitors: an overview with emphasis on pharmacokinetics and effects on oxidative drug metabolism. Clin Pharmacokinet 1997; 32 Suppl. 1: 1–21PubMedCrossRef Preskorn SH. Clinically relevant pharmacology of selective serotonin reuptake inhibitors: an overview with emphasis on pharmacokinetics and effects on oxidative drug metabolism. Clin Pharmacokinet 1997; 32 Suppl. 1: 1–21PubMedCrossRef
74.
go back to reference Fetell MR, Grossman SA, Fisher JD, et al. Preirradiation paclitaxel in glioblastoma multiforme: efficacy, pharmacology, and drug interactions. New approaches to brain tumor therapy central nervous system consortium. J Clin Oncol 1997; 15(9): 3121–8PubMed Fetell MR, Grossman SA, Fisher JD, et al. Preirradiation paclitaxel in glioblastoma multiforme: efficacy, pharmacology, and drug interactions. New approaches to brain tumor therapy central nervous system consortium. J Clin Oncol 1997; 15(9): 3121–8PubMed
75.
go back to reference Gilbert MR, Supko JG, Batchelor T, et al. Phase Iclinical and pharmacokinetic study of irinotecan in adults with recurrent malignant glioma. Clin Cancer Res 2003; 9(8): 2940–9PubMed Gilbert MR, Supko JG, Batchelor T, et al. Phase Iclinical and pharmacokinetic study of irinotecan in adults with recurrent malignant glioma. Clin Cancer Res 2003; 9(8): 2940–9PubMed
76.
go back to reference Prados MD, Yung WK, Wen PY, et al. Phase-1 trial of gefitinib and temozolomide in patients with malignant glioma: a North American Brain Tumor Consortium study. Cancer Chemother Pharmacol 2008; 61(6): 1059–67PubMedCrossRef Prados MD, Yung WK, Wen PY, et al. Phase-1 trial of gefitinib and temozolomide in patients with malignant glioma: a North American Brain Tumor Consortium study. Cancer Chemother Pharmacol 2008; 61(6): 1059–67PubMedCrossRef
77.
go back to reference Reardon DA, Quinn JA, Vredenburgh JJ, et al. Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma. Clin Cancer Res 2006; 12 (3 Pt 1): 860–8PubMedCrossRef Reardon DA, Quinn JA, Vredenburgh JJ, et al. Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma. Clin Cancer Res 2006; 12 (3 Pt 1): 860–8PubMedCrossRef
78.
go back to reference Relling MV, Hoffman JM. Should pharmacogenomic studies be required for new drug approval? Clin Pharmacol Ther 2007; 81(3): 425–8PubMedCrossRef Relling MV, Hoffman JM. Should pharmacogenomic studies be required for new drug approval? Clin Pharmacol Ther 2007; 81(3): 425–8PubMedCrossRef
79.
go back to reference Goetz MP, Kamal A, Ames MM. Tamoxifen pharmacogenomics: the role of CYP2D6 as a predictor of drug response. Clin Pharmacol Ther 2008; 83(1): 160–6PubMedCrossRef Goetz MP, Kamal A, Ames MM. Tamoxifen pharmacogenomics: the role of CYP2D6 as a predictor of drug response. Clin Pharmacol Ther 2008; 83(1): 160–6PubMedCrossRef
80.
go back to reference Kharasch ED, Walker A, Isoherranen N, et al. Influence of CYP3A5 genotype on the pharmacokinetics and pharmacodynamics of the cytochrome P4503A probes alfentanil and midazolam. Clin Pharmacol Ther 2007; 82(4): 410–26PubMedCrossRef Kharasch ED, Walker A, Isoherranen N, et al. Influence of CYP3A5 genotype on the pharmacokinetics and pharmacodynamics of the cytochrome P4503A probes alfentanil and midazolam. Clin Pharmacol Ther 2007; 82(4): 410–26PubMedCrossRef
81.
go back to reference Renbarger JL, McCammack KC, Rouse CE, et al. Effect of race on vincristineassociated neurotoxicity in pediatric acute lymphoblastic leukemia patients. Pediatr Blood Cancer 2008; 50(4): 769–71PubMedCrossRef Renbarger JL, McCammack KC, Rouse CE, et al. Effect of race on vincristineassociated neurotoxicity in pediatric acute lymphoblastic leukemia patients. Pediatr Blood Cancer 2008; 50(4): 769–71PubMedCrossRef
82.
go back to reference Renders L, Frisman M, Ufer M, et al. CYP3A5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients. Clin Pharmacol Ther 2007; 81(2): 228–34PubMedCrossRef Renders L, Frisman M, Ufer M, et al. CYP3A5 genotype markedly influences the pharmacokinetics of tacrolimus and sirolimus in kidney transplant recipients. Clin Pharmacol Ther 2007; 81(2): 228–34PubMedCrossRef
83.
go back to reference Xie HG, Wood AJ, Kim RB, et al. Genetic variability in CYP3A5 and its possible consequences. Pharmacogenomics 2004; 5(3): 243–72PubMedCrossRef Xie HG, Wood AJ, Kim RB, et al. Genetic variability in CYP3A5 and its possible consequences. Pharmacogenomics 2004; 5(3): 243–72PubMedCrossRef
84.
go back to reference Yasuda SU, Zhang L, Huang SM. The role of ethnicity in variability in response to drugs: focus on clinical pharmacology studies. Clin Pharmacol Ther 2008; 84(3): 417–23PubMedCrossRef Yasuda SU, Zhang L, Huang SM. The role of ethnicity in variability in response to drugs: focus on clinical pharmacology studies. Clin Pharmacol Ther 2008; 84(3): 417–23PubMedCrossRef
85.
go back to reference Dennison JB, Jones DR, Renbarger JL, et al. Effect of CYP3A5 expression on vincristine metabolism with human liver microsomes. J Pharmacol Exp Ther 2007; 321(2): 553–63PubMedCrossRef Dennison JB, Jones DR, Renbarger JL, et al. Effect of CYP3A5 expression on vincristine metabolism with human liver microsomes. J Pharmacol Exp Ther 2007; 321(2): 553–63PubMedCrossRef
86.
go back to reference Dennison JB, Kulanthaivel P, Barbuch RJ, et al. Selective metabolism of vincristine in vitro by CYP3A5. Drug Metab Dispos 2006; 34(8): 1317–27PubMedCrossRef Dennison JB, Kulanthaivel P, Barbuch RJ, et al. Selective metabolism of vincristine in vitro by CYP3A5. Drug Metab Dispos 2006; 34(8): 1317–27PubMedCrossRef
87.
go back to reference Dennison JB, Mohutsky MA, Barbuch RJ, et al. Apparent high CYP3A5 expression is required for significant metabolism of vincristine by human cryopreserved hepatocytes. J Pharmacol Exp Ther 2008; 327(1): 248–57PubMedCrossRef Dennison JB, Mohutsky MA, Barbuch RJ, et al. Apparent high CYP3A5 expression is required for significant metabolism of vincristine by human cryopreserved hepatocytes. J Pharmacol Exp Ther 2008; 327(1): 248–57PubMedCrossRef
88.
go back to reference Williams JA, Johnson K, Paulauskis J, et al. So many studies, too few subjects: establishing functional relevance of genetic polymorphisms on pharmaco-kinetics. J Clin Pharmacol 2006; 46(3): 258–64PubMedCrossRef Williams JA, Johnson K, Paulauskis J, et al. So many studies, too few subjects: establishing functional relevance of genetic polymorphisms on pharmaco-kinetics. J Clin Pharmacol 2006; 46(3): 258–64PubMedCrossRef
89.
go back to reference Proctor NJ, Tucker GT, Rostami-Hodjegan A. Predicting drug clearance from recombinantly expressed CYPs: intersystem extrapolation factors. Xenobiotica 2004; 34(2): 151–78PubMedCrossRef Proctor NJ, Tucker GT, Rostami-Hodjegan A. Predicting drug clearance from recombinantly expressed CYPs: intersystem extrapolation factors. Xenobiotica 2004; 34(2): 151–78PubMedCrossRef
Metadata
Title
A Quantitative Framework and Strategies for Management and Evaluation of Metabolic Drug-Drug Interactions in Oncology Drug Development
New Molecular Entities as Object Drugs
Authors
Dr Karthik Venkatakrishnan, PhD
Michael D. Pickard
Lisa L. von Moltke
Publication date
01-11-2010
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 11/2010
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.2165/11536740-000000000-00000

Other articles of this Issue 11/2010

Clinical Pharmacokinetics 11/2010 Go to the issue