577
Views
0
CrossRef citations to date
0
Altmetric
Review

Cytochrome P450 Enzymes: A Review on Drug Metabolizing Enzyme Inhibition Studies in Drug Discovery and Development

ORCID Icon, , & ORCID Icon
Pages 1355-1378 | Received 15 Jun 2021, Accepted 24 Aug 2021, Published online: 14 Sep 2021

References

  • Nelson DR , KamatakiT, WaxmanDJet al. The P450 superfamily: update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature. DNA Cell Biol.12(1), 1–51 (1993).
  • Rendic S , CarloFJD. Human cytochrome P450 enzymes: a status report summarising their reactions, substrates, inducers, and inhibitors. Drug Metab. Rev.29(1–2), 413–580 (1997).
  • Kumar GN , SurapaneniS. Role of drug metabolism in drug discovery and development. Med. Res. Rev.21(5), 397–411 (2001).
  • Kuzikov AV , MasamrekhRA, FilippovaTAet al. Electrochemical oxidation of estrogens as a method for CYP19A1 (aromatase) electrocatalytic activity determination. Electrochim. Acta.333, 135539 (2020).
  • Nainwal N . Recent advances in transcranial focused ultrasound (FUS) triggered brain delivery. Current Drug Targets18(11), 1225–1232 (2017).
  • Nettleton DO , EinolfHJ. Assessment of cytochrome p450 enzyme inhibition and inactivation in drug discovery and development. Curr. Top Med. Chem.11(4), 382–403 (2011).
  • Williams JA , HylandR, JonesBCet al. drug–drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab. Dispos.32(11), 1201–1208 (2004).
  • Lee J , ObachRS, FisherMB. Drug Metabolizing Enzymes: Cytochrome P450 and Other Enzymes in Drug Discovery and DevelopmentTaylor & Francis, FL, USA (2003).
  • Rao Gajula SN , ReddyGN, ReddyDS, SontiR. Pharmacokinetic drug–drug interactions: an insight into recent US FDA-approved drugs for prostate cancer. Bioanalysis12(22), 1647–1664 (2020).
  • Issa NT , WathieuH, OjoA, ByersWS, DakshanamurthyS. Drug metabolism in preclinical drug development: a survey of the discovery process, toxicology, and computational tools. Curr. Drug Metab.18(6), 556–565 (2017).
  • Pelkonen O , TurpeinenM, UusitaloJet al. Prediction of drug metabolism and interactions on the basis of in vitro investigations. Basic Clin. Pharmacol. Toxicol.96(3), 167–175 (2005).
  • Zhang Z , TangW. Drug metabolism in drug discovery and development. Acta Pharm. Sin. B.8(5), 721–732 (2018).
  • Roberts S . High-throughput screening approaches for investigating drug metabolism and pharmacokinetics. Xenobiotica31(8–9), 557–589 (2001).
  • Pradeep K , DileepR, PavanB. Importance of ADME and Bioanalysis in the Drug Discovery. J. Bioequiv. Availab.5, e31 (2013).
  • Palmer AM . New horizons in drug metabolism, pharmacokinetics and drug discovery. Drug News Perspect.16(1), 57–62 (2003).
  • Roberts S . High-throughput screening approaches for investigating drug metabolism and pharmacokinetics. Xenobiotica31(8–9), 557–589 (2001).
  • Yengi LG , LeungL, KaoJ. The evolving role of drug metabolism in drug discovery and development. Pharm. Res.24(5), 842 (2007).
  • Baillie TA . Metabolism and toxicity of drugs. Two decades of progress in industrial drug metabolism. Chem. Res. Toxicol.21(1), 129–137 (2008).
  • Lynch T , PriceAL. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am. Fam. Physician76(3), 391–396 (2007).
  • Weinshilboum R . Inheritance and drug response. N. Engl. J. Med.348(6), 529–537 (2003).
  • Zhang H , DavisCD, SinzMW, RodriguesAD. Cytochrome P450 reaction-phenotyping: an industrial perspective. N. Engl. J. Med.3(5), 667–687 (2007).
  • Rodrigues A , RushmoreTH. Cytochrome P450 pharmacogenetics in drug development: in vitro studies and clinical consequences. Curr. Drug Metab.3(3), 289–309 (2002).
  • Rodrigues AD . Integrated cytochrome P450 reaction phenotyping: attempting to bridge the gap between cDNA-expressed cytochromes P450 and native human liver microsomes. Biochem. Pharmacol.57(5), 465–480 (1999).
  • Lu AY , WangRW, LinJH. Cytochrome P450 in vitro reaction phenotyping: a re-evaluation of approaches used for P450 isoform identification. Drug Metab. Dispos.31(4), 345–350 (2003).
  • Williams JA , HurstSI, BaumanJet al. Reaction phenotyping in drug discovery: moving forward with confidence? Curr. Drug Metab. 4(6), 527–534 (2003).
  • Foye WO . Foye's Principles of Medicinal Chemistry.Lippincott Williams & Wilkins, PA, USA (2008).
  • Padmanabhan S . Handbook of Pharmacogenomics and Stratified Medicine.Elsevier Science, MA, USA (2014).
  • Lemke TL , WilliamsDA. Foye's Principles of Medicinal Chemistry.Wolters Kluwer Health/Lippincott Williams & Wilkins, PA, USA (2012).
  • Ueno K , SatoH. Sex-related differences in pharmacokinetics and pharmacodynamics of anti-hypertensive drugs. Hypertens. Res.35(3), 245–250 (2012).
  • McDonnell AM , DangCH. Basic review of the cytochrome p450 system. J. Adv. Pract. Oncol.4(4), 263 (2013).
  • Meyer UA . Pharmacogenetics and adverse drug reactions. J. Adv. Pract. Oncol.356(9242), 1667–1671 (2000).
  • Shenfield GM . Genetic polymorphisms, drug metabolism and drug concentrations. Clin. Biochem. Rev.25(4), 203 (2004).
  • Singh D , KashyapA, PandeyRV, SainiKS. Novel advances in cytochrome P450 research. Drug Discov. Today16(17–18), 793–799 (2011).
  • Yadav AS , ShahNR, CarlsonTJ, DriscollJP. Metabolite Profiling and Reaction Phenotyping for the in vitro Assessment of the Bioactivation of Bromfenac. Chem. Res. Toxicol.33(1), 249–257 (2019).
  • de Leon J , ArranzMJ, RuañoG. Pharmacogenetic testing in psychiatry: a review of features and clinical realities. Clin. Lab. Med.28(4), 599–617 (2008).
  • Li X , QuiggRJ, ZhouJ, GuW, RaoPN, ReedEF. Clinical utility of microarrays: current status, existing challenges and future outlook. Curr. Genomics9(7), 466–474 (2008).
  • US food and drug administration. RejRJCC. Center for devices and radiological health. 49(5), 840–841 (2003). http://www.fda.gov/cdrh
  • Yan Z , CaldwellGW. Metabolism profiling, and cytochrome P450 inhibition & induction in drug discovery. Curr. Top Med. Chem.1(5), 403–425 (2001).
  • Rodrigues AD . Drug–Drug Interactions.CRC Press, NY, USA (2019).
  • Kamel A , HarrimanS. Inhibition of cytochrome P450 enzymes and biochemical aspects of mechanism-based inactivation (MBI). Drug Discov. Today Technol.10(1), e177–e189 (2013).
  • Drexler DM , BelcastroJV, DickinsonKEet al. An automated high throughput liquid chromatography-mass spectrometry process to assess the metabolic stability of drug candidates. Assay Drug Dev. Technol.5(2), 247–264 (2007).
  • Youdim KA , SaundersKC. A review of LC-MS techniques and high-throughput approaches used to investigate drug metabolism by cytochrome P450s. J. Chromatogr. B.878(17–18), 1326–1336 (2010).
  • Li X , JiangG, LuoCet al. Ion trap array mass analyzer: structure and performance. Anal. Chem.81(12), 4840–4846 (2009).
  • Gajula SNR , NadimpalliN, SontiR. Drug metabolic stability in early drug discovery to develop potential lead compounds. Drug Metab. Rev.1–47 (2021). https://www.tandfonline.com/doi/abs/10.1080/03602532.2021.1970178
  • Kaufmann A , ButcherP, MadenK, WalkerS, WidmerM. Comprehensive comparison of liquid chromatography selectivity as provided by two types of liquid chromatography detectors (high resolution mass spectrometry and tandem mass spectrometry): “Where is the crossover point?”. Anal. Chim. Acta673(1), 60–72 (2010).
  • Kato H . Computational prediction of cytochrome P450 inhibition and induction. Drug Metab. Pharmacokinet.35(1), 30–44 (2020).
  • Tyzack JD , KirchmairJ. Computational methods and tools to predict cytochrome P450 metabolism for drug discovery. Chem. Biol. Drug Des.93(4), 377–386 (2019).
  • Daina A , MichielinO, ZoeteV. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep.7, 42717 (2017).
  • Hunt PA , SegallMD, TyzackJD. WhichP450: a multi-class categorical model to predict the major metabolizing CYP450 isoform for a compound. J. Comput. Aided Mol. Des.32(4), 537–546 (2018).
  • De Graaf C , VermeulenNP, FeenstraKA. Cytochrome P450 in silico: an integrative modeling approach. J. Pharmacol. Exp. Ther.48(8), 2725–2755 (2005).
  • Ring B , WrightonSA, MohutskyM. Reversible mechanisms of enzyme inhibition and resulting clinical significance. In: Enzyme Kinetics in Drug Metabolism.Springer, NJ, USA, 37–56 (2014).
  • Poli SM . Irreversible Cytochrome P450 Inhibition: Common Substructures and Implications for Drug Development.Wiley-VCH, Weinheim, Germany (2008).
  • Fontana E , DansetteP, PoliS. Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity. Curr. Drug Metab.6(5), 413–451 (2005).
  • Vaz RJ , KlabundeT, MannholdR, KubinyiH, FolkersG. Antitargets: Prediction and Prevention of Drug Side Effects.Wiley, KGaA, Weinheim (2008).
  • Kalgutkar AS , ObachRS, MaurerTS. Mechanism-based inactivation of cytochrome P450 enzymes: chemical mechanisms, structure-activity relationships and relationship to clinical drug–drug interactions and idiosyncratic adverse drug reactions. Curr. Drug Metab.8(5), 407–447 (2007).
  • Grimm SW , EinolfHJ, HallSDet al. The conduct of in vitro studies to address time-dependent inhibition of drug-metabolizing enzymes: a perspective of the pharmaceutical research and manufacturers of America. Drug Metab. Dispos.37(7), 1355–1370 (2009).
  • Rock D , SchragM, WienkersLC. Experimental characterisation of cytochrome P450 mechanism based inhibition. In: Handbook of Drug Metabolism.John Wiley & Sons, Inc., NJ, USA, 41–569 (2009).
  • Masimirembwa CM , ThompsonR, AnderssonTB. In vitro high throughput screening of compounds for favorable metabolic properties in drug discovery. Comb. Chem. High Throughput Screen.4(3), 245–263 (2001).
  • Ansede JH , ThakkerDR. High-throughput screening for stability and inhibitory activity of compounds toward cytochrome P450-mediated metabolism. J. Pharm. Sci.93(2), 239–255 (2004).
  • Zlokarnik G , GrootenhuisPDJ, WatsonJB. High throughput P450 inhibition screens in early drug discovery. Drug Discov. Today10(21), 1443–1450 (2005).
  • Lu C , DiL. In vitro and in vivo methods to assess pharmacokinetic drug–drug interactions in drug discovery and development. Biopharm. Drug Dispos.41(1–2), 3–31 (2020).
  • Schneider E , ClarkDS. Cytochrome P450 (CYP) enzymes and the development of CYP biosensors. Biosens. Bioelectron.39(1), 1–13 (2013).
  • Shumyantseva VV , MakhovaAA, ShikhEVet al. Bioelectrochemical systems as technologies for studying drug interactions related to cytochrome P450. Bionanoscience9(1), 79–86 (2019).
  • Fantuzzi A , CapriaE, MakLHet al. An electrochemical microfluidic platform for human P450 drug metabolism profiling. Anal. Chem.82(24), 10222–10227 (2010).
  • Shumyantseva VV , MakhovaAA, BulkoTVet al. Electrocatalytic cycle of P450 cytochromes: the protective and stimulating roles of antioxidants. RSC Adv.5(87), 71306–71313 (2015).
  • Sadeghi SJ , FantuzziA, GilardiG. Breakthrough in P450 bioelectrochemistry and future perspectives. Biochim. Biophys. Acta1814(1), 237–248 (2011).
  • Schneider E , ClarkDS. Cytochrome P450 (CYP) enzymes and the development of CYP biosensors. Biosens. Bioelectron.39(1), 1–13 (2013).
  • Di Nardo G , CastrignanòS, SadeghiSJ, BaravalleR, GilardiG. Bioelectrochemistry as a tool for the study of aromatisation of steroids by human aromatase. Electrochem. Commun.52, 25–28 (2015).
  • Kuzikov AV , MasamrekhRA, FilippovaTAet al. Electrochemical oxidation of estrogens as a method for CYP19A1 (aromatase) electrocatalytic activity determination. Electrochim. Acta333, 135539 (2020).
  • Nassar A-E , SchenkmanJ, RuslingJ. Faraday Transactions. Direct electron injection from electrodes to cytochrome P450 cam in biomembrane-like films. J. Chem. Soc.93(9), 1769–1774 (1997).
  • Shumyantseva VV , BulkoTV, KuzikovAVet al. All-electrochemical nanocomposite two-electrode setup for quantification of drugs and study of their electrocatalytical conversion by cytochromes P450. Electrochim. Acta336, 135579 (2020).
  • Jia L , LiuX. The conduct of drug metabolism studies considered good practice (II): in vitro experiments. Curr. Drug Metab.8(8), 822–829 (2007).
  • Lu C , LiAP. Enzyme Inhibition in Drug Discovery and Development: The Good and the Bad.Wiley, NJ, USA (2010).
  • Sowjanya G , GanapatyS, SharmaR. In vitro test methods for metabolite identification: a review. Asian J. Pharm. Pharmacol.5(3), 441–450 (2019).
  • Margolis JM , ObachRS. Impact of nonspecific binding to microsomes and phospholipid on the inhibition of cytochrome P4502D6: implications for relating in vitro inhibition data to in vivo drug interactions. Drug Metab. Dispos.31(5), 606–611 (2003).
  • Parmentier Y , BossantMJ, BertrandM, WaltherB. In Vitro Studies of Drug Metabolism. Comprehensive Medicinal Chemistry II5, 231–257 (2006).
  • Venkatakrishnan K , von MoltkeLL, GreenblattDJ. Human drug metabolism and the cytochromes P450: application and relevance of in vitro models. J. Clin. Pharmacol.41(11), 1149–1179 (2001).
  • Venkatakrishnan K , von MoltkeLL, CourtMH, HarmatzJS, CrespiCL, GreenblattDJ. Comparison between cytochrome P450 (CYP) content and relative activity approaches to scaling from cDNA-expressed CYPs to human liver microsomes: ratios of accessory proteins as sources of discrepancies between the approaches. Drug Metab. Dispos.28(12), 1493–1504 (2000).
  • Stringer RA , Strain-DamerellC, NicklinP, HoustonJB. Evaluation of recombinant cytochrome P450 enzymes as an in vitro system for metabolic clearance predictions. Drug Metab. Dispos.37(5), 1025–1034 (2009).
  • Brown HS , GaletinA, HallifaxD, HoustonJB. Prediction of in vivo drug–drug interactions from in vitro data. Clin. Pharmacokinet.45(10), 1035–1050 (2006).
  • Volpe DA , BalimanePV. Application of in vitro CYP and transporter assays to predict clinical drug–drug interactions. Bioanalysis10(9), 619–623 (2018).
  • Bjornsson TD , CallaghanJT, EinolfHJet al. The conduct of in vitro and in vivo drug–drug interaction studies: a PhRMA perspective. J. Clin. Pharmacol.43(5), 443–469 (2003).
  • Stresser DM , BlanchardAP, TurnerSDet al. Substrate-dependent modulation of CYP3A4 catalytic activity: analysis of 27 test compounds with four fluorometric substrates. Drug Metab. Dispos.28(12), 1440–1448 (2000).
  • Houston J , GaletinA. In Vitro Techniques to study drug–drug interactions of drug metabolism: cytochrome P450. 169–215 (2010).
  • Knights KM , StresserDM, MinersJO, CrespiCL. In vitro drug metabolism using liver microsomes. Curr. Protoc. Pharmacol.74(1), 7.8.1–7.8.24 (2016).
  • Wienkers LC , HeathTG. Predicting in vivo drug interactions from in vitro drug discovery data. Nat. Rev. Drug Discov.4(10), 825–833 (2005).
  • Fowler S , ZhangH. In vitro evaluation of reversible and irreversible cytochrome P450 inhibition: current status on methodologies and their utility for predicting drug–drug interactions. AAPS J.10(2), 410–424 (2008).
  • Obach RS , WalskyRL, VenkatakrishnanK, HoustonJB, TremaineLM. 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.78(6), 582–592 (2005).
  • Obach RS , WalskyRL, VenkatakrishnanK, HoustonJB, TremaineLM. 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.78(6), 582–592 (2005).
  • Obach RS , WalskyRL, VenkatakrishnanK, GamanEA, HoustonJB, TremaineLM. The utility of in vitro cytochrome P450 inhibition data in the prediction of drug–drug interactions. 316(1), 336–348 (2006).
  • Food U, Administration D. Guidance for industry: drug interaction studies – study design, data analysis, and implications for dosing and labeling. Center for Drug Evaluation and Research.US FDA, Silver Spring, MD, USA (2006). http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/DrugInteractionsLabeling/ucm093606.htm
  • Brown HS , ItoK, GaletinA, HoustonJB. Prediction of in vivo drug–drug interactions from in vitro data: impact of incorporating parallel pathways of drug elimination and inhibitor absorption rate constant. Br. J. Clin. Pharmacol.60(5), 508–518 (2005).
  • Riley RJ , GrimeK, WeaverR. Time-dependent CYP inhibition. Expert Opin. Drug Metab. Toxicol.3(1), 51–66 (2007).
  • Ghanbari F , Rowland-YeoK, BloomerJet al. A critical evaluation of the experimental design of studies of mechanism based enzyme inhibition, with implications for in vitro–in vivo extrapolation. Curr. Drug Metab.7(3), 315–334 (2006).
  • Silverman RB . Mechanism Based Enzyme InactiveCRC-Press, FL, USA (1988).
  • Mohutsky M , HallSD. Irreversible enzyme inhibition kinetics and drug–drug interactions. In: Enzyme Kinetics in Drug Metabolism.Springer, NY, USA, 57–91 (2014).
  • Obach RS , WalskyRL, VenkatakrishnanK. Mechanism-based inactivation of human cytochrome P450 enzymes and the prediction of drug–drug interactions. Drug Metab. Dispos.35(2), 246–255 (2007).
  • Atkinson A , KennyJR, GrimeK. Automated assessment of time-dependent inhibition of human cytochrome P450 enzymes using liquid chromatography-tandem mass spectrometry analysis. Drug Metab. Dispos.33(11), 1637–1647 (2005).
  • Parkinson A , KazmiF, BuckleyDBet al. An evaluation of the dilution method for identifying metabolism-dependent inhibitors of cytochrome P450 enzymes. Drug Metab. Dispos.39(8), 1370–1387 (2011).
  • Venkatakrishnan K , ObachR, Rostami-HodjeganA. Mechanism-based inactivation of human cytochrome P450 enzymes: strategies for diagnosis and drug–drug interaction risk assessment. Xenobiotica37(10–11), 1225–1256 (2007).
  • Burt HJ , GaletinA, HoustonJB. IC50-based approaches as an alternative method for assessment of time-dependent inhibition of CYP3A4. Xenobiotica40(5), 331–343 (2010).
  • Maurer TS , Tabrizi-FardMA, FungHL. Impact of mechanism-based enzyme inactivation on inhibitor potency: implications for rational drug discovery. J. Pharm. Sci.89(11), 1404–1414 (2000).
  • Berry LM , ZhaoZ. An examination of IC50 and IC50-shift experiments in assessing time-dependent inhibition of CYP3A4, CYP2D6 and CYP2C9 in human liver microsomes. Drug Metab. Lett.2(1), 51–59 (2008).
  • Yang J , JameiM, YeoKR, TuckerGT, Rostami-HodjeganA. Kinetic values for mechanism-based enzyme inhibition: assessing the bias introduced by the conventional experimental protocol. Eur. J. Pharm. Sci.26(3–4), 334–340 (2005).
  • Kent UM , RobertsES, ChunJ, HodgeK, JuncajJ, HollenbergPF. Inactivation of cytochrome P450 2E1 by tert-butylisothiocyanate. Chem. Res. Toxicol.11(10), 1154–1161 (1998).
  • Yang J , LiaoM, ShouMet al. Cytochrome p450 turnover: regulation of synthesis and degradation, methods for determining rates, and implications for the prediction of drug interactions. Curr. Drug Metab.9(5), 384–393 (2008).
  • Mayhew BS , JonesDR, HallSD. An in vitro model for predicting in vivo inhibition of cytochrome P450 3A4 by metabolic intermediate complex formation. Drug Metab. Dispos.28(9), 1031–1037 (2000).
  • Faber MS , FuhrU. Time response of cytochrome P450 1A2 activity on cessation of heavy smoking. Clin. Pharmacol. Ther.76(2), 178–184 (2004).
  • Venkatakrishnan K , ObachRS. In vitro-in vivo extrapolation of CYP2D6 inactivation by paroxetine: prediction of nonstationary pharmacokinetics and drug interaction magnitude. Drug Metab. Dispos.33(6), 845–852 (2005).
  • Greenblatt DJ , von MoltkeLL, HarmatzJSet al. Time course of recovery of cytochrome p450 3A function after single doses of grapefruit juice. Clin. Pharmacol. Ther.74(2), 121–129 (2003).
  • Rodrigues AD , LinJH. Screening of drug candidates for their drug–drug interaction potential. Curr. Opin. Chem. Biol.5(4), 396–401 (2001).
  • Ung YT , OngCE, PanY. Current high-throughput approaches of screening modulatory effects of xenobiotics on cytochrome P450 (CYP) enzymes. High-throughput7(4), 29 (2018).
  • Nayadu S , BeheraD, SharmaM, KaurG, GudiG. Fluorescent probe based CYP inhibition assay: a high throughput tool for early drug discovery screening. Rapid Commun. Mass Spectrom.5, 303–307 (2013).
  • Ansede JH , ThakkerDR. High-throughput screening for stability and inhibitory activity of compounds toward cytochrome P450-mediated metabolism. J. Pharm. Sci.93(2), 239–255 (2004).
  • Desai PK , TsengH, SouzaGR. Assembly of hepatocyte spheroids using magnetic 3D cell culture for CYP450 inhibition/induction. Int. J. Mol. Sci.18(5), 1085 (2017).
  • Ayrton J , PlumbR, LeavensW, MallettD, DickinsM, DearG. Application of a generic fast gradient liquid chromatography tandem mass spectrometry method for the analysis of cytochrome P450 probe substrates. Rapid Commun. Mass Spectrom.12(5), 217–224 (1998).
  • Zhang J-W , LiuY, LiW, HaoD-C, YangL. Inhibitory effect of medroxyprogesterone acetate on human liver cytochrome P450 enzymes. Eur. J. Clin. Pharmacol.62(7), 497–502 (2006).
  • Jin C , HeX, ZhangFet al. Inhibitory mechanisms of celastrol on human liver cytochrome P450 1A2, 2C19, 2D6, 2E1 and 3A4. Xenobiotica45(7), 571–577 (2015).
  • de Man FM , GoeyAKL, van SchaikRHN, MathijssenRHJ, BinsS. Individualization of irinotecan treatment: a review of pharmacokinetics, pharmacodynamics, and pharmacogenetics. Clin. Pharmacokinet.57(10), 1229–1254 (2018).
  • Hanioka N , OzawaS, JinnoHet al. Interaction of irinotecan (CPT-11) and its active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) with human cytochrome P450 enzymes. Drug Metab. Dispos.30(4), 391–396 (2002).
  • Lin HL , KentUM, HollenbergPF. Mechanism-based inactivation of cytochrome P450 3A4 by 17 alpha-ethynylestradiol: evidence for heme destruction and covalent binding to protein. J. Pharmacol. Exp. Ther.301(1), 160–167 (2002).
  • Stanczyk FZ , ArcherDF, BhavnaniBR. Ethinyl estradiol and 17β-estradiol in combined oral contraceptives: pharmacokinetics, pharmacodynamics and risk assessment. Contracept.87(6), 706–727 (2013).
  • Wang K , FengX, ChaiL, CaoS, QiuF. The metabolism of berberine and its contribution to the pharmacological effects. Drug Metab. Rev.49(2), 139–157 (2017).
  • Guo Y , LiF, MaX, ChengX, ZhouH, KlaassenCD. CYP2D plays a major role in berberine metabolism in liver of mice and humans. Xenobiotica.41(11), 996–1005 (2011).
  • Kim HG , LeeHS, JeonJSet al. Quasi-Irreversible Inhibition of CYP2D6 by Berberine. Pharmaceutics.12(10), 916 (2020).

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.