1,242
Views
37
CrossRef citations to date
0
Altmetric
Review

Reaction phenotyping to assess victim drug-drug interaction risks

Pages 1105-1115 | Received 14 Feb 2017, Accepted 10 Aug 2017, Published online: 18 Aug 2017

References

  • Nicolas J-M, Espie P, Molimard M. Gender and interindividual variability in pharmacokinetics. Drug Metab Rev. 2009;41(3):408–421.
  • Soldin OP, Chung SH, Mattison DR. Sex differences in drug disposition. J Biomed Biotechnol. 2011;187103:14.
  • Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14.
  • Sansone-Parsons A, Krishna G, Simon J, et al. Effects of age, gender, and race/ethnicity on the pharmacokinetics of posaconazole in healthy volunteers. Antimicrob Agents Chemother. 2007;51(2):495–502.
  • Morgan ET. Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clin Pharmacol Ther. 2009;85(4):434–438.
  • Blot SI, Pea F, Lipman J. The effect of pathophysiology on pharmacokinetics in the critically ill patient - Concepts appraised by the example of antimicrobial agents. Adv Drug Delivery Rev. 2014;77:3–11.
  • Roden DM, George AL Jr. The genetic basis of variability in drug responses. Nat Rev Drug Discovery. 2002;1(1):37–44.
  • Kirchheiner J, Roots I, Goldammer M, et al. Effect of genetic polymorphisms in cytochrome P450 (CYP) 2C9 and CYP2C8 on the pharmacokinetics of oral antidiabetic drugs: clinical relevance. Clin Pharmacokinet. 2005;44(12):1209–1225.
  • Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138(1):103–141.
  • [Cited 2017 Aug 15]. Available from: https://www.certara.com/software/pbpk-modeling-and-simulation/. SIMCYP Substrate File.
  • [Cited 2017 Aug 15]. Available from: http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm115979.htm.
  • [Cited 2017 Aug 15]. Available from: http://www.fda.gov/downloads/Drugs/Guidance-ComplianceRegulatoryInformation/Guidances/ucm292362.pdf.
  • Pinto N, Dolan ME. Clinically relevant genetic variations in drug metabolizing enzymes. Curr Drug Metab. 2011;12(5):487–497.
  • Joffe HV, Xu R, Johnson FB, et al. Warfarin dosing and cytochrome P450 2C9 polymorphisms. Thromb Haemost. 2004;91(6):1123–1128.
  • Flora DR, Rettie AE, Brundage RC, et al. CYP2C9 genotype-dependent warfarin pharmacokinetics: impact of CYP2C9 genotype on R- and S-warfarin and their oxidative metabolites. J Clin Pharmacol. 2017;57(3):382–393.
  • Proctor NJ, Tucker GT, Rostami-Hodjegan A. Predicting drug clearance from recombinantly expressed CYPs: intersystem extrapolation factors. Xenobiotica. 2004;34(2):151–178.
  • Venkatakrishnan K, von Moltke LL, Greenblatt DJ. Relative quantities of catalytically active CYP 2C9 and 2C19 in human liver microsomes: application of the relative activity factor approach. J Pharm Sci. 1998;87(7):845–853.
  • Venkatakrishnan K, Von Moltke LL, Greenblatt DJ. Application of the relative activity factor approach in scaling from heterologously expressed cytochromes P450 to human liver microsomes: studies on amitriptyline as a model substrate. J Pharmacol Exp Ther. 2001;297(1):326–337.
  • Stormer E, Von Moltke LL, Greenblatt DJ. Scaling drug biotransformation data from cDNA-expressed cytochrome P-450 to human liver: a comparison of relative activity factors and human liver abundance in studies of mirtazapine metabolism. J Pharmacol Exp Ther. 2000;295(2):793–801.
  • Crespi CL, Miller VP. The use of heterologously expressed drug metabolizing enzymes- state of the art and prospects for the future. Pharmacol Ther. 1999;84(2):121–131.
  • Galetin A, Brown C, Hallifax D, et al. Utility of recombinant enzyme kinetics in prediction of human clearance: impact of variability, CYP3A5, and CYP2C19 on CYP3A4 probe substrates. Drug Metab Dispos. 2004;32(12):1411–1420.
  • Emoto C, Murase S, Iwasaki K. Approach to the prediction of the contribution of major cytochrome P450 enzymes to drug metabolism in the early drug-discovery stage. Xenobiotica. 2006;36(8):671–683.
  • Gibson CR, Lu P, Maciolek C, et al. Using human recombinant UDP-glucuronosyltransferase isoforms and a relative activity factor approach to model total body clearance of laropiprant (MK-0524) in humans. Xenobiotica. 2013;43(12):1027–1036.
  • Youdim KA, Zayed A, Dickins M, et al. Application of CYP3A4 in vitro data to predict clinical drug-drug interactions; predictions of compounds as objects of interaction. Br J Clin Pharmacol. 2008;65(5):680–692.
  • Siu YA, Lai WG. Impact of probe substrate selection on cytochrome P450 reaction phenotyping using the relative activity factor. Drug Metab Dispos. 2017;45(2):183–189.
  • Chen Y, Liu L, Nguyen K, et al. Utility of intersystem extrapolation factors in early reaction phenotyping and the quantitative extrapolation of human liver microsomal intrinsic clearance using recombinant cytochromes P450. Drug Metab Dispos. 2011;39(3):373–382.
  • Yang X, Atkinson K, Di L. Novel cytochrome P450 reaction phenotyping for low-clearance compounds using the hepatocyte relay method. Drug Metab Dispos. 2016;44(3):460–465.
  • Zhang H, Davis CD, Sinz MW, et al. Cytochrome P450 reaction-phenotyping: an industrial perspective. Expert Opin Drug Metab Toxicol. 2007;3(5):667–687.
  • Kazmi F, Barbara JE, Yerino P, et al. A long-standing mystery solved: the formation of 3-hydroxydesloratadine is catalyzed by CYP2C8 but prior glucuronidation of desloratadine by UDP-glucuronosyltransferase 2B10 is an obligatory requirement. Drug Metab Dispos. 2015;43(4):523–533.
  • Kazmi F, Yerino P, Ogilvie BW, et al. Assessment under initial rate conditions of the selectivity and time course of cytochrome P450 inactivation in pooled human liver microsomes and hepatocytes: optimization of inhibitor conditions used for reaction phenotyping studies. 19th North American ISSX Meeting, Poster; 2014; San Francisco, CA.
  • Li C, Kalyanaraman N. Reaction phenotyping. In: Zhang D, Surapaneni S, editors. ADME-enabling technologies in drug design and development. Hoboken, NJ: Wiley; 2012. p. 189–212.
  • Hyland R, Jones BC, Smith DA. Identification of the cytochrome P450 enzymes involved in the N-oxidation of voriconazole. Drug Metab Dispos. 2003;31(5):540–547.
  • Bohnert T, Patel A, Templeton I, et al. Evaluation of a new molecular entity as a victim of metabolic drug-drug interactions-an industry perspective. Drug Metab Dispos. 2016;44(8):1399–1423.
  • Lapham K, Baumann JN, Walsky RL, et al. Digoxin and tranilast identified as novel isoform-selective inhibitors of human UDP-glucuronosyltransferase 1A9 (UGT1A)) activity (Poster P108). Drug Metab Rev. 2012;44(S1):82.
  • Zientek MA, Youdim K. Reaction phenotyping: advances in the experimental strategies used to characterize the contribution of drug-metabolizing enzymes. Drug Metab Dispos. 2015;43(1):163–181.
  • Jinno N, Tagashira M, Tsurui K, et al. Contribution of cytochrome P450 and UDT-glucuronosyltransferase to the metabolism of drugs containing carboxylic acid groups: risk assessment of acylglucuronides using human hepatocytes. Xenobiotica. 2014;44(8):677–686.
  • Ghosal A, Yuan Y, Tong W, et al. Characterization of human liver enzymes involved in the biotransformation of boceprevir, a hepatitis C virus protease inhibitor. Drug Metab Dispos. 2011;39(3):510–521.
  • Siu M, Johnson TO, Wang Y, et al. N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: discovery of PF-915275. Bioorg Med Chem Lett. 2009;19(13):3493–3497.
  • Brodney MA, Beck EM, Butler CR, et al. Utilizing Structures of CYP2D6 and BACE1 complexes to reduce risk of drug-drug interactions with a novel series of centrally efficacious BACE1 inhibitors. J Med Chem. 2015;58(7):3223–3252.
  • Hurst S, Williams JA, Hansel S. Reaction phenotyping. In Drug MetabDrug Design Deve., Edited by Donglu Zhang, Mingshe Zhu, William G. Humphreys, John Wiley & Son, NJ, 2008;477–512.
  • Nijenhuis CM, Schellens JHM, Beijnen JH. Regulatory aspects of human radiolabeled mass balance studies in oncology: concise review. Drug Metab Rev. 2016;48(2):266–280.
  • Tse S, Leung L, Raje S, et al. Disposition and metabolic profiling of [14C]cerlapirdine using accelerator mass spectrometry. Drug Metab Dispos. 2014;42(12):2023–2032.
  • Dowty ME, Lin J, Ryder TF, et al. The pharmacokinetics, metabolism, and clearance mechanisms of tofacitinib, a janus kinase inhibitor, in humans. Drug Metab Dispos. 2014;42(4):759–773.
  • Young Graeme C, Seymour M. Application of (14)C-accelerator MS in pharmaceutical development. Bioanalysis. 2015;7(5):513–517.
  • Bowers GD, Tenero D, Patel P, et al. Disposition and metabolism of GSK2251052 in humans: a novel boron-containing antibiotic. Drug Metab Dispos. 2013;41(5):1070–1081.
  • Dave M, Nash M, Young GC, et al. Disposition and metabolism of darapladib, a lipoprotein-associated phospholipase A2 inhibitor, in humans. Drug Metab Dispos. 2014;42(3):415–430.
  • Johnson TR, Tan W, Goulet L, et al. Metabolism, excretion and pharmacokinetics of [14C]crizotinib following oral administration to healthy subjects. Xenobiotica. 2015;45(1):45–59.
  • Xu H, O’Gorman M, Tan W, et al. The effects of ketoconazole and rifampin on the single-dose pharmacokinetics of crizotinib in healthy subjects. Eur J Clin Pharmacol. 2015;71(12):1441–1449.
  • Fahmi OA, Hurst S, Plowchalk D, et al. Comparison of different algorithms for predicting clinical drug-drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds as precipitants of interaction. Drug Metab Dispos. 2009;37(8):1658–1666.
  • Shou M, Hayashi M, Pan Y, et al. Modeling, prediction, and in vitro in vivo correlation of CYP3A4 induction. Drug Metab Dispos. 2008;36(11):2355–2370.
  • 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):837–844.
  • Gibbs JP, Hyland R, Youdim K. Minimizing polymorphic metabolism in drug discovery: evaluation of the utility of in vitro methods for predicting pharmacokinetic consequences associated with CYP2D6 metabolism. Drug Metab Dispos. 2006;34(9):1516–1522.
  • Boulenc X, Djebli N, Shi J, et al. Effects of omeprazole and genetic polymorphism of CYP2C19 on the clopidogrel active metabolite. Drug Metab Dispos. 2012;40(1):187–197.
  • Di L, Obach RS. Addressing the challenges of low clearance in drug research. AAPS Journal. 2015;17(2):352–357.
  • Di L, Trapa P, Obach RS, et al. A novel relay method for determining low-clearance values. Drug Metab Dispos. 2012;40(9):1860–1865.
  • Di L, Atkinson K, Orozco CC, et al. In vitro-in vivo correlation for low-clearance compounds using hepatocyte relay method. Drug Metab Dispos. 2013;41(12):2018–2023.
  • Chan TS, Yu H, Moore A, et al. Meeting the challenge of predicting hepatic clearance of compounds slowly metabolized by cytochrome P450 using a novel hepatocyte model, HepatoPac. Drug Metab Dispos. 2013;41(12):2024–2032.
  • Lin C, Shi J, Moore A, et al. Prediction of drug clearance and drug-drug interactions in microscale cultures of human hepatocytes. Drug Metab Dispos. 2016;44(1):127–136.
  • Bonn B, Svanberg P, Janefeldt A, et al. Determination of human hepatocyte intrinsic clearance for slowly metabolized compounds: comparison of a primary hepatocyte/stromal cell co-culture with plated primary hepatocytes and HepaRG. Drug Metab Dispos. 2016;44(4):527–533.
  • Vivares A, Salle-Lefort S, Arabeyre-Fabre C, et al. Morphological behaviour and metabolic capacity of cryopreserved human primary hepatocytes cultivated in a perfused multiwell device. Xenobiotica. 2015;45(1):29–44.
  • Schaefer M, Schanzle G, Bischoff D, et al. Upcyte human hepatocytes: a potent in vitro tool for the prediction of hepatic clearance of metabolically stable compounds. Drug Metab Dispos. 2016;44(3):435–444.
  • Godoy P, Hewitt NJ, Albrecht U, et al. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol. 2013;87(8):1315–1530.
  • Scotcher D, Billington S, Brown J, et al. Microsomal and cytosolic scaling factors in dog and human kidney cortex and application for in vitro-in vivo extrapolation of renal metabolic clearance. Drug Metab Dispos. 2017;45(5):556–568.
  • Masuo Y, Nagamori S, Hasegawa A, et al. Utilization of liver microsomes to estimate hepatic intrinsic clearance of monoamine oxidase substrate drugs in humans. Pharm Res. 2017;34(6):1233–1243.
  • Leung L, Yang X, Strelevitz Timothy J, et al. Clearance prediction of targeted covalent inhibitors by in vitro-in vivo extrapolation of hepatic and extrahepatic clearance mechanisms. Drug Metab Dispos. 2017;45(1):1–7.
  • Zhang Z-Y, Wong YN. Enzyme kinetics for clinically relevant CYP inhibition. Curr Drug Metab. 2005;6(3):241–257.
  • Fowler S, Guerini E, Qiu N, et al. Low potential of basimglurant to be involved in drug-drug interactions: influence of non-Michaelis-Menten P450 kinetics on fraction metabolized. J Pharmacol Exp Ther. 2017;360(1):164–173.
  • Wu B. Substrate inhibition kinetics in drug metabolism reactions. Drug Metab Rev. 2011;43(4):440–456.
  • Von Moltke LL, Greenblatt DJ, Grassi JM, et al. Multiple human cytochromes contribute to biotransformation of dextromethorphan in-vitro: role of CYP2C9, CYP2C19, CYP2D6, and CYP3A. J Pharm Pharmacol. 1998;50(9):997–1004.
  • Andersson T, Miners JO, Veronese ME, et al. Diazepam metabolism by human liver microsomes is mediated by both S-mephenytoin hydroxylase and CYP3A isoforms. Br J Clin Pharmacol. 1994;38(2):131–137.
  • Fowler S, Kletzl H, Finel M, et al. A UGT2B10 splicing polymorphism common in african populations may greatly increase drug exposure. J Pharmacol Exp Ther. 2015;352(2):358–367.
  • Hutzler JM, Tracy TS. Atypical kinetic profiles in drug metabolism reactions. Drug Metab Dispos. 2002;30(4):355–362.
  • Tracy TS. Atypical enzyme kinetics: their effect on in vitro-in vivo pharmacokinetic predictions and drug interactions. Curr Drug Metab. 2003;4(5):341–346.
  • Tracy TS, Hummel MA. Modeling kinetic data from in vitro drug metabolism enzyme experiments. Drug Metab Rev. 2004;36(2):231–242.
  • Shou M, Lin Y, Lu P, et al. Enzyme kinetics of cytochrome P450-mediated reactions. Curr Drug Metab. 2001;2(1):17–36.
  • Korzekwa KR, Krishnamachary N, Shou M, et al. Evaluation of atypical cytochrome P450 kinetics with two-substrate models: evidence that multiple substrates can simultaneously bind to cytochrome P450 active sites. Biochemistry. 1998;37(12):4137–4147.
  • Houston JB, Galetin A. Modelling atypical CYP3A4 kinetics: principles and pragmatism. Arch Biochem Biophys. 2005;433(2):351–360.
  • Houston JB, Kenworthy KE. In vitro-in vivo scaling of CYP kinetic data not consistent with the classical Michaelis-Menten model. Drug Metab Dispos. 2000;28(3):246–254.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.