958
Views
50
CrossRef citations to date
0
Altmetric
Review

Drug metabolism and metabolite safety assessment in drug discovery and development

&
Pages 1071-1085 | Received 12 Jan 2018, Accepted 31 Aug 2018, Published online: 14 Sep 2018

References

  • Yengi LG, Leung L, Kao J. The evolving role of drug metabolism in discovery and development. Pharm Res. 2007;24:842–858.
  • Zhu MS, Zhang DL, Humphreys WG. Drug metabolism in drug discovery and development. New Jersey: Wiley; 2007.
  • Mascitti V, Stevens BD, Choi C, et al. Design and evaluation of a 2-(2, 3, 6-trifluorophenyl) acetamide derivative as an agonist of the GPR119 receptor. Bioorg Med Chem Lett. 2011;21:1306–1309.
  • Stepan AF, Mascitti V, Beaumont K, et al. Metabolism-guided drug design. Med Chem Commun. 2013;4:631–652.
  • Fura A. Role of pharmacologically active metabolites in drug discovery and development. Drug Discov Today. 2006;11:133–142.
  • Ling KH, Leeson GA, Burmaster SD, et al. Metabolism of terfenadine associated with CYP3A4 activity in human hepatic microsomes. Drug Metab Dispos. 1995;23:631–636.
  • Yun CH, Okerholm RA, Guengerich EP. Oxidation of the antihistamine drug terfenadine in human liver microsomes: role of cytochrome P450 3A (4) in N-dealkylation and C-hydroxylation. Drug Metab Dispos. 1993;21:403–409.
  • Roy ML, Dumaine R, Brown AM. hERG, a primary human ventricular target of the nonsedating antihistamine terfenadine. Circulation. 1996;94:817–823.
  • Honig PK, Wortham DC, Zamani K, et al. Terfenadine-ketoconazole interaction. JAMA. 1993;269:1513–1518.
  • FDA. Guidance for Industry: Safety testing of drug metabolites. 2016.
  • ICH. Guidance for industry: M3 (R2) nonclinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals. 2010.
  • ICH. Guidance for industry: M3 (R2) nonclinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals: questions and answers (R2). 2013.
  • Wan H. What ADME tests should be conducted for preclinical studies? ADMET & DMPK. 2013;1:19–28.
  • Pagliarusco S, Martinucci S, Bordini E, et al. Tissue distribution and characterization of drug-related material in rats and dogs after repeated oral administration of casopitant. Drug Metab Dispos. 2011;39:283–293.
  • Pellegatti M, Pagliarusco S. Drug and metabolite concentrations in tissues in relationship to tissue adverse findings: a review. Expert Opin Drug Metab Toxicol. 2011;7:137–146.
  • Timmerman P, Blech S, White S, et al. Best practices for metabolite quantification in drug development: updated recommendation from the European Bioanalysis Forum. Bioanalysis. 2016;8:1297–1305.
  • Yu HB, Bischoff D, Tweedie D. Challenges and solutions to metabolites in safety testing: impact of the international conference on harmonization M3(R2) guidance. Expert Opin Drug Metab Toxicol. 2010;6:1539–1549.
  • Ma SG, Chowdhury SK. Analytical strategies for assessment of human metabolites in preclinical safety testing. Anal Chem. 2011;83:5028–5036.
  • Yeung CK, Fujioka Y, Hachad H, et al. Are circulating metabolites important in drug-drug interactions? quantitative analysis of risk prediction and inhibitory potency. Clin Pharmacol Ther. 2011;89:105–113.
  • FDA. Guidance for industry: drug interaction studies — study design, data analysis, implications for dosing, and labeling recommendations. 2012.
  • ICH. Guideline on the investigation of drug interactions. 2012.
  • Callegari E, Kalgutkar AS, Leung L, et al. Drug metabolites as cytochrome P450 inhibitors: a retrospective analysis and proposed algorithm for evaluation of the pharmacokinetic interaction potential of metabolites in drug discovery and development. Drug Metab Dispos. 2013;41:2047–2055.
  • Yu HB, Balani SK, Chen WC, et al. Contribution of metabolites to P450 inhibition-based drug-drug interactions: scholarship from the drug metabolism leadership group of the innovation and quality consortium metabolite group. Drug Metab Dispos. 2015;43:620–630.
  • Yu HB, Tweedie D. A perspective on the contribution of metabolites to drug-drug interaction potential: the need to consider both circulating levels and inhibition potency. Drug Metab Dispos. 2013;41:536–540.
  • Zetterberg C, Maltais F, Laitinen L, et al. VX-509 (decernotinib)-mediated CYP3A time-dependent inhibition: an aldehyde oxidase metabolite as a perpetrator of drug-drug interactions. Drug Metab Dispos. 2016;44:1286–1295.
  • Tornio A, Filppula AM, Kailari O, et al. Glucuronidation converts clopidogrel to a strong time-dependent inhibitor of CYP2C8: a phase II metabolite as a perpetrator of drug–drug interactions. Clin Pharmacol Ther. 2014;96:498–507.
  • Itkonen MK, Tornio A, Neuvonen M, et al. Clopidogrel markedly increases plasma concentrations of CYP2C8 substrate pioglitazone. Drug Metab Dispos. 2016;44:1364–1371.
  • Leung L, Kalgutkar AS, Obach RS. Metabolic activation in drug-induced liver injury. Drug Metab Rev. 2012;44:18–33.
  • Kalgutkar AS, Fate G, Didiuk MT, et al. Toxicophores, reactive metabolites and drug safety: when is it a cause for concern? Expert Rev Clin Pharmacol. 2008;1:515–531.
  • Park BK, Boobis A, Clarke S, et al. Managing the challenge of chemically reactive metabolites in drug develop. Nat Rev Drug Discov. 2011;10:292–306.
  • Stachulski AV, Baillie TA, Park BK, et al. The generation, detection, and effects of reactive drug metabolites. Med Res Rev. 2013;33:985–1080.
  • Brink A, Pähler A, Funk C, et al. Minimizing the risk of chemically reactive metabolite formation of new drug candidates: implications for preclinical drug design. Drug Discov Today. 2017;22:751–756.
  • Lammert C, Bjornsson E, Niklasson A, et al. Oral medications with significant hepatic metabolism at higher risk for hepatic adverse events. Hepatology. 2010;51:615–620.
  • Gómez-Lechóna MJ, Tolosaa L, Donatoa T. Metabolic activation and drug-induced liver injury: in vitro approaches for the safety risk assessment of new drugs. J Appl Toxicol. 2016;36:752–768.
  • Aleo MD, Luo Y, Swiss R, et al. Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump. Hepatology. 2014;60:1015–1022.
  • Liver Tox. A website on drug-induced liver injury. [cited 2017 May 18]. Available from: http://livertox.nlm.nih.gov.
  • Sistare FD, Mattes WB, LeCluyse EL. The promise of new technologies to reduce, refine, or replace animal use while reducing risks of drug induced liver injury in pharmaceutical development. ILAR J. 2016;57:186–211.
  • Onakpoya IJ, Heneghan CJ, Aronson JK. Worldwide withdrawal of medicinal products because of adverse drug reactions: a systematic review and analysis. Crit Rev Toxicol. 2016;46:477–489.
  • Farmer PB. DNA and protein adducts as markers of genotoxicity. Toxicol Lett. 2004;149:3–9.
  • Liu X, Pisha E, Tonetti DA, et al. Antiestrogenic and DNA damaging effects induced by tamoxifen and toremifene metabolites. Chem Res Toxicol. 2003;16:832–837.
  • Kolbanovskiy A, Kuzmin V, Shastry A, et al. Base selectivity and effects of sequence and DNA secondary structure on the formation of covalent adducts derived from the equine estrogen metabolite 4-hydroxyequilenin. Chem Res Toxicol. 2005;18:1737–1747.
  • Gómez-Lechón MJ, Lahoz A, Gombau L, et al. In vitro evaluation of potential hepatotoxicity induced by drugs. Curr Pharm Des. 2010;16:1963–1977.
  • Stepan AF, Walker DP, Bauman J, et al. Structural alert/reactive metabolite concept as applied in medicinal chemistry to mitigate the risk of idiosyncratic drug toxicity: a perspective based on the critical examination of trends in the top 200 drugs marketed in the United States. Chem Res Toxicol. 2011;24:1345–1410.
  • Kalgutkar AS, Gardner I, Obach RS, et al. A comprehensive listing of bioactivation pathways of organic functional groups. Curr Drug Metab. 2005;6:161–225.
  • Erve JC, Maynard JW Jr, Tonn G, et al. Bioactivation of sitaxentan in liver microsomes, hepatocytes, and expressed human P450s with characterization of the glutathione conjugate by liquid chromatography tandem mass spectrometry. Chem ResToxicol. 2013;26:926–936.
  • Kim JH, Hoi WG, Lee S, et al. Revisiting the metabolism and bioactivation of ketoconazole in human and mouse using liquid chromatography-mass spectrometry-based metabolomics. Int J Mol Sci. 2017;18:621–639.
  • Cirello AL, Dumouchel JL, Gunduz M, et al. In vitro ocular metabolism and bioactivation of ketoconazole in rat, rabbit and human. Drug Metab Pharmacokinet. 2017;32:121–126.
  • Bessone F, Hernandez N, Roma MG, et al. Hepatotoxicity induced by coxibs: how concerned should we be? Expert Opin Drug Saf. 2016;15:1463–1475.
  • Bauman JN, Frederick KS, Sawant A, et al. Comparison of the bioactivation potential of the antidepressant and hepatotoxin nefazodone with aripiprazole, a structural analog and marketed drug. Drug Metab Dispos. 2008;36:1016–1029.
  • Foster AJ, Prime LH, Gustafsson F, et al. Bioactivation of the cannabinoid receptor antagonist rimonabant to a cytotoxic iminium ion metabolite. Chem Res Toxicol. 2013;26:124–135.
  • Li X, Zhong K, Guo Z1, et al. Fasiglifam (TAK-875) inhibits hepatobiliary transporters: a possible factor contributing to fasiglifam-induced liver injury. Drug Metab Dispos. 2015;43:1751–1759.
  • Skonberg C, Olsen J, Madsen KG, et al. Metabolic activation of carboxylic acids. Expert Opin Drug Metab Toxicol. 2008;4:425–438.
  • Miyashita T, Kimura K, Fukami T, et al. Evaluation and mechanistic analysis of the cytotoxicity of the acyl glucuronide of nonsteroidal anti-inflammatory drugs. Drug Metab Dispos. 2014;42:1–8.
  • Ebner T, Wagner K, Wienen W. Dabigatran acylglucuronide, the major human metabolite of dabigatran: in vitro formation, stability, and pharmacological activity. Drug Metab Dispos. 2010;38:1567–1575.
  • Regan SL, Maggs JL, Hammond TG, et al. Acyl glucuronides: the good, the bad and the ugly. Biopharm Drug Dispos. 2010;31:367–395.
  • Sawamura R, Okudaira N, Watanabe K, et al. Predictability of idiosyncratic drug toxicity risk for carboxylic acid-containing drugs based on the chemical stability of acyl glucuronide. Drug Metab Dispos. 2010;38:1857–1864.
  • Banoglu E. Current status of the cytosolic sulfotransferases in the metabolic activation of promutagens and procarcinogens. Curr Drug Metab; 2000;1:1–30.
  • Zhang XC, Sharma AM, Uetrecht J. Identification of danger signals in nevirapine-induced skin rash. Chem Res Toxicol. 2013;26:410–421.
  • Wen B, Chen Y, Fitch WL. Metabolic activation of nevirapine in human liver microsomes: dehydrogenation and inactivation of cytochrome P450 3A4. Drug Metab Dispos. 2009;37:1557–1562.
  • Diao XX, Pang XY, Xie C, et al. Bioactivation of 3-n-butylphthalide via sulfation of its major metabolite 3-hydroxy-NBP: mediated mainly by sulfotransferase 1A1s. Drug Metab Dispos. 2014;42:774–781.
  • Sharma AM, Li Y, Novalen M, et al. Bioactivation of nevirapine to a reactive quinone methide: implications for liver injury. Chem Res Toxicol. 2012;25:1708–1719.
  • Dekker SJ, Zhang Y, Vos JC, et al. Different reactive metabolites of nevirapine require distinct glutathione S-transferase isoforms for bioinactivation. Chem Res Toxicol. 2016;29:2136–2144.
  • Sharma AM, Klarskov K, Uetrecht J. Nevirapine bioactivation and covalent binding in the skin. Chem Res Toxicol. 2013;26:410–421.
  • Sharma AM, Novalen M, Tanino T, et al. 12-OH-nevirapine sulfate, formed in the skin, is responsible for nevirapine-induced skin rash. Chem Res Toxicol. 2013;26:817–827.
  • Diao X, Deng P, Xie C, et al. Metabolism and pharmacokinetics of 3-n-butylphthalide (NBP) in humans: the role of cytochrome P450s and alcohol dehydrogenase in biotransformation. Drug Metab Dispos. 2013;41:430–444.
  • Thompson RA, Isin EM, Ogese MO, et al. Reactive metabolites: current and emerging risk and hazard assessments. Chem Res Toxicol. 2016;29:505–533.
  • Claesson A, Spjuth A. On mechanism of reactive metabolite formation from drugs. Mini Rev Med Chem. 2013;13:720–729.
  • Obach RS, Kalgutkar AS, Ryder TF, et al. In vitro metabolism and covalent binding of enolcarboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: insights into the hepatotoxicity of sudoxicam. Chem Res Toxicol. 2008;21:1890–1899.
  • Grillo MP. Detecting reactive drug metabolites for reducing the potential for drug toxicity. Expert Opin Drug Metab Toxicol. 2015;11:1281–1302.
  • Kalgutkar AS. Liabilities associated with the formation of hard electrophiles in reactive metabolite trapping screens. Chem Res Toxicol. 2017;30:220–238.
  • Ho HK, Chan JC, Hardy KD, et al. Mechanism-based inactivation of CYP450 enzymes: a case study of lapatinib. Drug Metab Rev. 2015;47:21–28.
  • Iwamura A, Nakajima M, Oda S, et al. Toxicological potential of acyl glucuronides and its assessment. Drug Metab Pharmacokinet. 2017;32:2–11.
  • Chen Z, Holt TG, Pivnichny JV, et al. A simple in vitro model to study the stability of acylglucuronides. J Pharmacol Toxicol Methods. 2007;55:91–95.
  • Iwamura A, Ito M, Mitsui H, et al. Toxicological evaluation of acyl glucuronides utilizing half-lives, peptide adducts, and immunostimulation assays. Toxicol in Vitro. 2015;30:241–249.
  • Jinno N, Ohashi S, Agashira M, et al. A simple method to evaluate reactivity of acylglucuronides optimized for early stage drug discovery. Biol Pharm Bull. 2013;36:1509–1513.
  • Zhong S, Jones R, Lu W, et al. A new rapid in vitro assay for assessing reactivity of acyl glucuronides. Drug Metab Dispos. 2015;43:1711–1717.
  • Lammert C, Einarsson S, Saha C, et al. Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology. 2008;47:2003–2009.
  • Dalvie D, Kang P, Ziente M, et al. Effect of intestinal glucuronidation in limiting hepatic exposure and bioactivation of raloxifene in humans and rats. Chem Res Toxicol. 2008;21:2260–2271.
  • Waring MJ. Lipophilicity in drug discovery. Expert Opin Drug Discov. 2010;5:235–248.
  • Arnott JA, Planey SL. The influence of lipophilicity in drug discovery and design. Expert Opin Drug Discov. 2012;7:863–875.
  • Meanwell NA. Improving drug candidates by design: a focus on physicochemical properties as a means of improving compound disposition and safety. Chem Res Toxicol. 2011;24:1420–1456.
  • Price DA, Blagg J, Jones L, et al. Physicochemical drug properties associated with in vivo toxicological outcomes: a review. Expert Opin Drug Metab Toxicol. 2009;5:921–931.
  • Hughes JD, Blagg J, Price DA, et al. Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorg Med Chem Lett. 2008;18:4872–4875.
  • Chen MJ, Borlak J, Tong WD. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology. 2013;58:388–396.
  • Utkarsh D, Loretz C, Li A. In vitro evaluation of hepatotoxic drugs in human hepatocytes from multiple donors: identification of P450 activity as a potential risk factor for drug-induced liver injuries. Chem Biol Interact. 2016;255:12–22.
  • Takimoto T, Kijima T, Otani Y, et al. Polymorphisms of CYP2D6 gene and gefitinib-induced hepatotoxicity. Clin Lung Cancer. 2013;14:502–507.
  • Perwitasari DA, Atthobari J, Wilffert B. Pharmacogenetics of isoniazid-induced hepatotoxicity. Drug Metab Rev. 2015;47:222–228.
  • Fukunaga K, Nakagawa H, Ishikawa T, et al. ABCB1 polymorphism is associated with atorvastatin-induced liver injury in Japanese population. BMC Genetics. 2016;17:79–85.
  • Chen R, Wang J, Tang SW, et al. Association of polymorphisms in drug transporter genes (SLCO1B1 and SLC10A1) and anti-tuberculosis drug-induced hepatotoxicity in a Chinese cohort. Tuberculosis. 2015;95:68–74.
  • Baillie TA. Approaches to the assessment of stable and chemically reactive drug metabolites in early clinical trials. Chem Res Toxicol. 2009;22:263–266.
  • Dalvie D, Kalgutkar AS, Chen W. Practical approaches to resolving reactive metabolite liabilities in early discovery. Drug Metab Rev. 2015;47:56–70.
  • Kalgutkar AS, Dalvie D. Predicting toxicities of reactive metabolite-positive drug candidates. Annu Rev Pharmacol Toxicol. 2015;55:35–54.
  • Tang W, Lu AYH. Metabolic bioactivation and drug-related adverse effects: current status and future directions from a pharmaceutical research perspective. Drug Metab Rev. 2010;42:225–249.
  • Martignoni M, Groothuis GM, de Kanter R. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol. 2006;2:875–894.
  • Argika UA, Potter PM, Hutzler JM, et al. Challenges and opportunities with non-CYP enzymes aldehyde oxidase, carboxylesterase, and UDP-glucuronosyltransferase: focus on reaction, phenotyping and prediction of human clearance. The AAPS J. 2016;18:1391–1405.
  • Preissner SC, Hoffmann MF, Preissner R, et al. Polymorphic cytochrome P450 enzymes (CYPs) and their role in personalized therapy. PLoS One. 2013;8:e82562.
  • Baillie TA, Rettie AE. Role of biotransformation in drug-induced toxicity: influence of intra- and inter-species differences in drug metabolism. Drug Metab Pharmacokinet. 2011;26:15–29.
  • Naidoo P, Chetty VV, Chetty M. Impact of CYP polymorphisms, ethnicity and sex differences in metabolism on dosing strategies: the case of efavirenz. Eur J Clin Pharmacol. 2014;70:379–389.
  • De KR, Monshouwer M, Draaisma AL. Prediction of whole-body metabolic clearance of drugs through the combined use of slices from rat liver, lung, kidney, small intestine and colon. Xenobiotica. 2004;34:229–241.
  • Bell LC, Wang JL, Probe ADM. E and test hypotheses: a PATH beyond clearance in vitro-in vivo correlations in early drug discovery. Expert Opin Drug Metab Toxicol. 2012;8:1131–1155.
  • Levy G, Yacobi A. Letter: effect of plasma protein binding on elimination of warfarin. J Pharm Sci. 1974;63:805–806.
  • Lin X, Skolnik S, Chen X, et al. Attenuation of intestinal absorption by major efflux transporters: quantitative tools and strategies using a Caco-2 model. Drug Metab Dispos. 2011;39:265–274.
  • Chu X, Bleasby K, Evers R. Species differences in drug transporters and implications for translating preclinical findings to humans. Expert Opin Drug Metab Toxicol. 2013;9:237–252.
  • Van Vleet TR, Liu H, Lee A, et al. Acyl glucuronide metabolites: implications for drug safety assessment. Toxicol Lett. 2017;272:1–7.
  • Wen B, Fitch W. Analytical strategies for the screening and evaluation of chemically reactive drug metabolites. Expert Opin Drug Metab Toxicol. 2009;5:39–54.

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.