Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-r6qrq Total loading time: 0 Render date: 2024-04-25T13:38:24.458Z Has data issue: false hasContentIssue false

6 - Reliability of reactive metabolite and covalent binding assessments in prediction of idiosyncratic drug toxicity

from I - SPECIFIC AREAS OF PREDICTIVE TOXICOLOGY

Published online by Cambridge University Press:  06 December 2010

Jinghai J. Xu
Affiliation:
Merck Research Laboratory, New Jersey
Laszlo Urban
Affiliation:
Novartis Institutes for Biomedical Research, Massachusetts
Get access

Summary

INTRODUCTION

Safety-related attrition continues to be a major concern in the pharmaceutical industry. Of a total of 548 drugs approved in the period from 1975 to 1999, 45 drugs (8.2 percent) acquired 1 or more black box warnings and 16 (2.9 percent) were withdrawn from the market owing to idiosyncratic adverse drug reactions (IADRs) that were not predicted from animal testing and/or clinical trials. IADRs (also known as type B ADRs) are unrelated to known drug pharmacology, and are generally dose-independent. Because the frequency of occurrence of IADRs is very low (1 in 10,000 to 1 in 100,000), these reactions are often not detected until the drug has gained broad exposure in a large patient population. Importantly, standard regulatory animal toxicity studies have traditionally shown a poor concordance with occurrence of IADRs in humans. Life-threatening IADRs noted for drugs include hepatotoxicity, severe cutaneous reactions, aplastic anemia, and blood dyscrasias. Recognizing these issues, many pharmaceutical companies are increasing their efforts in implementing predictive in vitro tools to identify potential safety liabilities earlier in the drug discovery process so that they can be eliminated via chemical intervention or the compound suspended from further development. One component of such assays is aimed at understanding a drug candidate's propensity to undergo reactive metabolite formation.

LINKING METABOLISM WITH TOXICITY

Drugs are metabolized via oxidative, reductive, and hydrolytic pathways (phase I reactions), which lead to a modest increase in aqueous solubility; phase II conjugations modify the newly introduced functionality to form O- and N-glucuronides, sulfate and acetate esters, all with increased hydrophilicity relative to the unconjugated metabolite.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Kramer, JA, Sagartz, JE, Morris, DL. The application of discovery toxicology and pathology towards the design of safer pharmaceutical lead candidates. Nat Rev Drug Discov. 2007;6(8):636–649.CrossRefGoogle ScholarPubMed
Lasser, KE, Allen, PD, Woolhandler, SJ, et al. Timing of new black box warnings and withdrawals for prescription medications. JAMA. 2002;287(17):2215–2220.CrossRefGoogle ScholarPubMed
Olson, H, Betton, G, Robinson, D, et al. Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol. 2000;32(1):56–67.CrossRefGoogle ScholarPubMed
Miller, EC, Miller, JA. Mechanisms of chemical carcinogenesis: nature of proximate carcinogens and interactions with macromolecules. Pharmacol Rev. 1966;18(1):805–838.Google ScholarPubMed
Mitchell, JR, Jollow, DJ, Potter, WZ, et al. Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism. J Pharmacol Exp Ther. 1973;187(1):185–194.Google ScholarPubMed
Jollow, DJ, Mitchell, JR, Potter, WZ, et al. Acetaminophen-induced hepatic necrosis. II. Role of covalent binding in vivo. J Pharmacol Exp Ther. 1973;187(1):195–202.Google ScholarPubMed
Potter, WZ, Davis, DC, Mitchell, JR, et al. Acetaminophen-induced hepatic necrosis. III. Cytochrome P-450-mediated covalent binding in vitro. J Pharmacol Exp Ther. 1973;187(1):203–210.Google Scholar
Mitchell, JR, Jollow, D, Potter, WZ, et al. Acetaminophen-induced hepatic necrosis. IV. Protective role of glutathione. J Pharmacol Exp Ther. 1973;187(1):211–217.Google ScholarPubMed
Dahlin, DC, Miwa, GT, Lu, AY, et al. N-acetyl-p-benzoquinone imine: A cytochrome P-450-mediated oxidation product of acetaminophen. Proc Natl Acad Sci USA. 1984;81(5):1327–1331.CrossRefGoogle ScholarPubMed
Zhao, Z, Baldo, BA, Rimmer, Jbeta-Lactam allergenic determinants: fine structural recognition of a cross-reacting determinant on benzylpenicillin and cephalothin. Clin Exp Allergy. 2002;32(11):1644–1650.CrossRefGoogle ScholarPubMed
Hess, DA, Sisson, ME, Suria, H, et al. Cytotoxicity of sulfonamide reactive metabolites: Apoptosis and selective toxicity of CD8(+) cells by the hydroxylamine of sulfamethoxazole. FASEB J. 1999:13(13):1688–1698.CrossRefGoogle ScholarPubMed
Lecoeur, S, Andre, C, Beaune, PH. Tienilic acid-induced autoimmune hepatitis: Anti-liver and-kidney microsomal type 2 autoantibodies recognize a three-site conformational epitope on cytochrome P4502C9. Mol Pharmacol. 1996;50(2):326–333.Google ScholarPubMed
Bourdi, M, Gautier, JC, Mircheva, J, et al. Anti-liver microsomes autoantibodies and dihydralazine-induced hepatitis: Specificity of autoantibodies and inductive capacity of the drug. Mol Pharmacol. 1992;42(2):280–285.Google ScholarPubMed
Bourdi, M, Chen, W, Peter, RM, et al. Human cytochrome P450 2E1 is a major autoantigen associated with halothane hepatitis. Chem Res Toxicol. 1996;9(7):1159–1166.CrossRefGoogle ScholarPubMed
Kalgutkar, AS, Soglia, JR. Minimising the potential for metabolic activation in drug discovery. Expert Opin Drug Metab Toxicol. 2005;1(1):91–142.CrossRefGoogle ScholarPubMed
Kalgutkar, AS, Gardner, I, Obach, RS, et al. A comprehensive listing of bioactivation pathways of organic functional groups. Curr Drug Metab. 2005;6(3):161–225.CrossRefGoogle ScholarPubMed
Correia, MA, Krowech, G, Caldera-Munoz, P, et al. Morphine metabolism revisited. II. Isolation and chemical characterization of a glutathionylmorphine adduct from rat liver microsomal preparations. Chem Biol Interact. 1984;51(1):13–24.CrossRefGoogle ScholarPubMed
Soglia, JR, Harriman, SP, Zhao, S, et al. The development of a higher throughput reactive intermediate screening assay incorporating micro-bore liquid chromatography-micro-electrospray ionization-tandem mass spectrometry and glutathione ethyl ester as an in vitro conjugating agent. J Pharm Biomed Anal. 2004;36(1):105–116.CrossRefGoogle Scholar
Baillie, TA, Davis, MR. Mass spectrometry in the analysis of glutathione conjugates. Biol Mass Spectrom. 1993;22(6):319–325.CrossRefGoogle ScholarPubMed
Dieckhaus, CM, Fernández-Metzler, CL, King, R, et al. Negative ion tandem mass spectrometry for the detection of glutathione conjugates. Chem Res Toxicol. 2005;18(4):630–638.CrossRefGoogle ScholarPubMed
Mutlib, A, Lam, W, Atherton, J, et al. Application of stable isotope labeled glutathione and rapid scanning mass spectrometers in detecting and characterizing reactive metabolites. Rapid Commun Mass Spectrom. 2005;19(23):3482–3492.CrossRefGoogle ScholarPubMed
Gan, J, Harper, TW, Hsueh, MM, et al. Dansyl glutathione as a trapping agent for the quantitative estimation and identification of reactive metabolites. Chem Res Toxicol. 2005;18(5):896–903.CrossRefGoogle ScholarPubMed
Soglia, JR, Contillo, LG, Kalgutkar, AS, et al. A semiquantitative method for the determination of reactive metabolite conjugate levels in vitro utilizing liquid chromatography-tandem mass spectrometry and novel quaternary ammonium glutathione analogues. Chem Res Toxicol. 2006;19(3):480–490.CrossRefGoogle ScholarPubMed
Sahali-Sahly, Y, Balani, SK, Lin, JH, et al. In vitro studies on the metabolic activation of the furanopyridine L-754,394, a highly potent and selective mechanism-based inhibitor of cytochrome P450 3A4. Chem Res Toxicol. 1996;9(6):1007–1012.CrossRefGoogle ScholarPubMed
Olsen, R, Molander, P, Øvrebø, S, et al. Reaction of glyoxal with 2'-deoxyguanosine, 2'-deoxyadenosine, 2'-deoxycytidine, cytidine, thymidine, and calf thymus DNA: identification of DNA adducts. Chem Res Toxicol. 2005;18(4):730–739.CrossRefGoogle ScholarPubMed
Gorrod, JW, Whittlesea, CM, Lam, SP. Trapping of reactive intermediates by incorporation of 14C-sodium cyanide during microsomal oxidation. Adv Exp Med Biol. 1991;283:657–664.CrossRefGoogle ScholarPubMed
Argoti, D, Liang, L, Conteh, A, et al. Cyanide trapping of iminium ion reactive intermediates followed by detection and structure identification using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Chem Res Toxicol. 2005;18(10):1537–1544.CrossRefGoogle Scholar
Walker, GS, Atherton, J, Bauman, J, et al. Determination of degradation pathways and kinetics of acyl glucuronides by NMR spectroscopy. Chem Res Toxicol. 2007;20(6):876–886.CrossRefGoogle ScholarPubMed
Johnson, CH, Wilson, ID, Harding, JR, et al. NMR spectroscopic studies on the in vitro acyl glucuronide migration kinetics of Ibuprofen ((+/-)-(R,S)-2-(4-isobutylphenyl) propanoic acid), its metabolites, and analogues. Anal Chem. 2007;79(22):8720–8727.CrossRefGoogle Scholar
Ding, A, Ojingwa, JC, McDonagh, AF, et al. Evidence for covalent binding of acyl glucuronides to serum albumin via an imine mechanism as revealed by tandem mass spectrometry. Proc Natl Acad Sci USA. 1993;90(9):3797–3801.CrossRefGoogle ScholarPubMed
Benet, LZ, Spahn-Langguth, H, Iwakawa, S, et al. Predictability of the covalent binding of acidic drugs in man. Life Sci. 1993;53(8):PL141–146.CrossRefGoogle ScholarPubMed
Evans, DC, Watt, AP, Nicoll-Griffith, DA, et al. Drug-protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development. Chem Res Toxicol. 2004;17(1):3–16.CrossRefGoogle ScholarPubMed
Zhang, D, Ogan, M, Gedamke, R, et al. Protein covalent binding of maxipost through a cytochrome P450-mediated ortho-quinone methide intermediate in rats. Drug Metab Dispos. 2003;31(7):837–845.CrossRefGoogle ScholarPubMed
Zhang, D, Krishna, R, Wang, L, et al. Metabolism, pharmacokinetics, and protein covalent binding of radiolabeled MaxiPost (BMS-204352) in humans. Drug Metab Dispos. 2005;33(1):83–93.CrossRefGoogle ScholarPubMed
Jurva, U, Wikstrom, HV, Weidolf, L, et al. Comparison between electrochemistry/mass spectrometry and cytochrome P450 catalyzed oxidation reactions. Rapid Commun Mass Spectrom. 2003;17(8):800–810.CrossRefGoogle ScholarPubMed
Johansson, T, Weidolf, L, Jurva, U. Mimicry of phase I drug metabolism – Novel methods for metabolite characterization and synthesis. Rapid Commun Mass Spectrom. 2007;21(14):2323–2331.CrossRefGoogle ScholarPubMed
Getek, TA, Korfmacher, WA, McRae, TA, et al. Utility of solution electrochemistry mass spectrometry for investigating the formation and detection of biologically important conjugates of acetaminophen. J Chromatogr. 1989;474(1):245–256.CrossRefGoogle ScholarPubMed
Jurva, U, Holmen, A, Gronberg, G, et al. Electrochemical generation of electrophilic drug metabolites: Characterization of amodiaquine quinoneimine and cysteinyl conjugates by MS, IR, and NMR. Chem Res Toxicol. 2008;21(4):928–935.CrossRefGoogle ScholarPubMed
Smith, KS, Smith, PL, Heady, TN, et al. In vitro metabolism of tolcapone to reactive intermediates: Relevance to tolcapone liver toxicity. Chem Res Toxicol. 2003;16(2):123–128.CrossRefGoogle ScholarPubMed
Madsen, KG, Skonberg, C, Jurva, U, et al. Bioactivation of diclofenac in vitro and in vivo: Correlation to electrochemical studies. Chem Res Toxicol. 2008;21(5):1107–1119.CrossRefGoogle ScholarPubMed
Madsen, KG, Olsen, J, Skonberg, C, et al. Development and evaluation of an electrochemical method for studying reactive phase-I metabolites: Correlation to in vitro drug metabolism. Chem Res Toxicol. 2007;20(5):821–831.CrossRefGoogle ScholarPubMed
Madsen, KG, Gronberg, G, Skonberg, C, et al. Electrochemical oxidation of troglitazone: Identification and characterization of the major reactive metabolite in liver microsomes. Chem Res Toxicol. 2008;21(10):2035–2041.CrossRefGoogle ScholarPubMed
Larrey, D, Tinel, M, Amouyal, G, et al. Genetically determined oxidation polymorphism and drug hepatotoxicity. Study of 51 patients. J Hepatol. 1989;8(2):158–164.CrossRefGoogle ScholarPubMed
Olanow, CW. Tolcapone and hepatotoxic effects. Tasmar advisory panel. Arch Neurol. 2000;57(2)263–267.CrossRefGoogle ScholarPubMed
Aithal, GP. Diclofenac-induced liver injury: A paradigm of idiosyncratic drug toxicity. Expert Opin Drug Saf. 2004;3(6):519–523.CrossRefGoogle ScholarPubMed
Schulte, PF. Risk of clozapine-associated agranulocytosis and mandatory white blood cell monitoring. Ann Pharmacother. 2006;40(4):683–688.CrossRefGoogle ScholarPubMed
Burt, RA. Review of adverse reactions associated with cinoxacin and other drugs used to treat urinary tract infections. Urology. 1984;23(1):101–107.CrossRefGoogle ScholarPubMed
Faich, GA, Moseley, RH. Troglitazone (Rezulin) and hepatic injury. Pharmacoepidemiol Drug Saf. 2001;30(2):155–160.Google Scholar
O'Neill, PM, Harrison, AC, Storr, RC, et al. The effect of fluorine substitution on the metabolism and antimalarial activity of amodiaquine. J Med Chem. 1994;37(9): 1362–1370.CrossRefGoogle Scholar
Tang, W. The metabolism of diclofenac – Enzymology and toxicology perspectives. Curr Drug Metab. 2003;4(4):319–329.CrossRefGoogle Scholar
Liu, ZC, Uetrecht, JP. Clozapine is oxidized by activated human neutrophils to a reactive nitrenium ion that irreversibly binds to the cells. J Pharmacol Exp Ther. 1995;275(3):1476–1483.Google ScholarPubMed
Uetrecht, JP. Metabolism of clozapine by neutrophils. Possible implications for clozapine-induced agranulocytosis. Drug Saf. 1992;7(Suppl 1):51–56.CrossRefGoogle ScholarPubMed
Lai, WG, Zahid, N, Uetrecht, JP. Metabolism of trimethoprim to a reactive iminoquinone methide by activated human neutrophils and hepatic microsomes. J Pharmacol Exp Ther. 1999;291(1):292–299.Google ScholarPubMed
Kassahun, K, Pearson, PG, Tang, W, et al. Studies on the metabolism of troglitazone to reactive intermediates in vitro and in vivo. Evidence for novel biotransformation pathways involving quinone methide formation and thiazolidinedione ring scission. Chem Res Toxicol. 2001;14(1):62–70.CrossRefGoogle Scholar
Alvarez-Sanchez, R, Montavon, F, Hartung, T, et al. Thiazolidinedione bioactivation: A comparison of the bioactivation potentials of troglitazone, rosiglitazone, and pioglitazone using stable isotope-labeled analogues and liquid chromatography tandem mass spectrometry. Chem Res Toxicol. 2006;19(8):1106–1116.CrossRefGoogle ScholarPubMed
Baughman, TM, Graham, RA, Wells-Knecht, K, et al. Metabolic activation of pioglitazone identified from rat and human liver microsomes and freshly isolated hepatocytes. Drug Metab Dispos. 2005;33(6):733–738.CrossRefGoogle ScholarPubMed
Cruciani, G, Carosati, E, Boeck, B, et al. MetaSite: Understanding metabolism in human cytochromes from the perspective of the chemist. J Med Chem. 2005;48(22):6970–6979.CrossRefGoogle ScholarPubMed
Trunzer, M, Faller, B, Zimmerlin, A. Metabolic soft spot identification and compound optimization in early discovery phases using MetaSite and LC-MS/MS validation. J Med Chem. 2009;52(2):329–335.CrossRefGoogle Scholar
Ahlström, MM, Ridderström, M, Zamora, I, et al. CYP2C9 structure-metabolism relationships: Optimizing the metabolic stability of COX-2 inhibitors. J Med Chem. 2007;50(18):4444–4452.CrossRefGoogle ScholarPubMed
Boyer, D, Bauman, JN, Walker, DP, et al. Utility of MetaSite in improving metabolic stability of the neutral indomethacin amide derivative and selective cyclooxygenase-2 (COX-2) inhibitor 2-(1-(4-chlorobenzoyl)-5-methoxy-2-methyl-1H-indol-3-yl]-N-phenethyl-acetamide. Drug Metab Dispos. 2009;37(5):999–1008.CrossRefGoogle ScholarPubMed
Remmel, RP, Crews, BC, Kozak, KR, et al. Studies on the metabolism of the novel, selective cyclooxygenase-2 inhibitor indomethacin phenethylamide in rat, mouse, and human liver microsomes: Identification of active metabolites. Drug Metab Dispos. 2004;32(1):113–122.CrossRefGoogle Scholar
Koymans, L, Lenthe, JH, Straat, R, et al. A theoretical study on the metabolic activation of paracetamol by cytochrome P-450: Indications for a uniform oxidation mechanism. Chem Res Toxicol. 1989;2(1):60–66.CrossRefGoogle ScholarPubMed
Koymans, L, Lenthe, JH, Donné-op Den Kelder, GM, et al. Mechanisms of activation of phenacetin to reactive metabolites by cytochrome P-450: A theoretical study involving radical intermediates. Mol Pharmacol. 1990;37(3):452–460.Google ScholarPubMed
Erve, JC, Svensson, MA, Euler-Chelpin, H, et al. Characterization of glutathione conjugates of the remoxipride hydroquinone metabolite NCQ-344 formed in vitro and detection following oxidation by human neutrophils. Chem Res Toxicol. 2004;17(4):564–571.CrossRefGoogle ScholarPubMed
Kalgutkar, AS, Vaz, AD, Lame, ME, et al. Bioactivation of the nontricyclic antidepressant nefazodone to a reactive quinone-imine species in human liver microsomes and recombinant cytochrome P450 3A4. Drug Metab Dispos. 2005;33(2):243–253.CrossRefGoogle ScholarPubMed
Dearden, JC. In silico prediction of drug toxicity. J Comput Aided Mol Des. 2003;17(2–4):119–127.CrossRefGoogle ScholarPubMed
Kalgutkar, AS, Dalvie, DK, Aubrecht, J, et al. Genotoxicity of 2-(3-chlorobenzyloxy)-6-(piperazinyl)pyrazine, a novel 5-hydroxytryptamine2c receptor agonist for the treatment of obesity: Role of metabolic activation. Drug Metab Dispos. 2007;35(6):848–858.CrossRefGoogle ScholarPubMed
Singh, R, Silva Elipe, MV, Pearson, PG, et al. Metabolic activation of a pyrazinone-containing thrombin inhibitor. Evidence for novel biotransformation involving pyrazinone ring oxidation, rearrangement, and covalent binding to proteins. Chem Res Toxicol. 2003;16(2):198–207.CrossRefGoogle ScholarPubMed
Amos, HE, Lake, BG, Artis, J. Possible role of antibody specific for a practolol metabolite in the pathogenesis of oculomucocutaneous syndrome. Br Med J. 1978;1(6110):402–404.CrossRefGoogle ScholarPubMed
Orton, C, Lowery, C. Practolol metabolism. III. Irreversible binding of [14C]practolol metabolite(s) to mammalian liver microsomes. J Pharmacol Exp Ther. 1981;219(1):207–212.Google Scholar
Borchard, U. Pharmacokinetics of beta-adrenoceptor blocking agents: Clinical significance of hepatic and/or renal clearance. Clin Physiol Biochem. 1990;8(Suppl 2):28–34.Google ScholarPubMed
Gardner, I, Leeder, JS, Chin, T, et al. A comparison of the covalent binding of clozapine and olanzapine to human neutrophils in vitro and in vivo. Mol Pharmacol. 1998;53(6):999–1008.Google ScholarPubMed
Uetrecht, J, Zahid, N, Tehim, A, et al. Structural features associated with reactive metabolite formation in clozapine analogues. Chem Biol Interact. 1997;104(2–3)117–129.CrossRefGoogle ScholarPubMed
Goldkind, L, Laine, L. A systematic review of NSAIDs withdrawn from the market due to hepatotoxicity: Lessons learned from the bromfenac experience. Pharmacoepidemiol Drug Saf. 2006;15(4):213–220.CrossRefGoogle ScholarPubMed
Wang, J, Davis, M, Li, F, et al. A novel approach for predicting acyl glucuronide reactivity via Schiff base formation: Development of rapidly formed peptide adducts for LC/MS/MS measurements. Chem Res Toxicol. 2004;17(9):1206–1216.CrossRefGoogle ScholarPubMed
Yardley-Jones, A, Anderson, D, Parke, DV. The toxicity of benzene and its metabolism and molecular pathology in human risk assessment. Br J Ind Med. 1991;48(7):437–444.Google ScholarPubMed
Rietjens, IM, Besten, C, Hanzlik, RP, et al. Cytochrome P450-catalyzed oxidation of halobenzene derivatives. Chem Res Toxicol. 1997;10(6):629–635.CrossRefGoogle ScholarPubMed
Jacobsen, W, Kuhn, B, Soldner, A, et al. Lactonization is the critical first step in the disposition of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor atorvastatin. Drug Metab Dispos. 2000;28(11):1369–1378.Google ScholarPubMed
Prueksaritanont, T, Subramanian, R, Fang, X, et al. Glucuronidation of statins in animals and humans: A novel mechanism of statin lactonization. Drug Metab Dispos. 2002;30(5):505–512.CrossRefGoogle ScholarPubMed
Savi, P, Pereillo, JM, Uzabiaga, MF, et al. Identification and biological activity of the active metabolite of clopidogrel. Thromb Haemost. 2000;84(5):891–896.CrossRefGoogle ScholarPubMed
Savi, P, Zachayus, JL, Delesque-Touchard, N, et al. The active metabolite of Clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts. Proc Natl Acad Sci USA. 2006;103(29):11069–11074.CrossRefGoogle ScholarPubMed
Herbert, JM, Savi, P. P2Y12, a new platelet ADP receptor, target of clopidogrel. Semin Vasc Med. 2003;3(2):113–122.Google ScholarPubMed
Olbe, L, Carlsson, E, Lindberg, P. A proton-pump inhibitor expedition: The case histories of omeprazole and esomeprazole. Nat Rev Drug Discov. 2003;2(2):132–139.CrossRefGoogle ScholarPubMed
Fellenius, E, Berglindh, T, Sachs, G, et al. Substituted benzimidazoles inhibit gastric acid secretion by blocking (H+ + K+)ATPase. Nature. 1981;290(5802):159–161.CrossRefGoogle Scholar
Durand, A, Thenot, JP, Bianchetti, G, et al. Comparative pharmacokinetic profile of two imidazopyridine drugs: Zolpidem and alpidem. Drug Metab Rev. 1992;24(2):239–266.CrossRefGoogle ScholarPubMed
Chen, Q, Ngui, JS, Doss, GA, et al. Cytochrome P450 3A4-mediated bioactivation of raloxifene: Irreversible enzyme inhibition and thiol adduct formation. Chem Res Toxicol. 2002;15(7):907–914.CrossRefGoogle ScholarPubMed
Kemp, DC, Fan, PW, Stevens, JC. Characterization of raloxifene glucuronidation in vitro: Contribution of intestinal metabolism to presystemic clearance. Drug Metab Dispos. 2002;30(6):694–700.CrossRefGoogle ScholarPubMed
Rashed, MS, Streeter, AJ, Nelson, SD. Investigations of the N-hydroxylation of 3'-hydroxyacetanilide, a non-hepatotoxic potential isomer of acetaminophen. Drug Metab Dispos. 1989;17(4):355–359.Google Scholar
Masubuchi, N, Makino, C, Murayama, N. Prediction of in vivo potential for metabolic activation of drugs into chemically reactive intermediate: Correlation of in vitro and in vivo generation of reactive intermediates and in vitro glutathione conjugate formation in rats and humans. Chem Res Toxicol. 2007;20(3):455–464.CrossRefGoogle ScholarPubMed
Obach, RS, Kalgutkar, AS, Soglia, JR, et al. Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from nonhepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose. Chem Res Toxicol. 2008;21(9):1814–1822.CrossRefGoogle ScholarPubMed
Bauman, JN, Kelly, JM, Tripathy, S, et al. Can in vitro metabolism-dependent covalent binding data distinguish hepatotoxic from nonhepatotoxic drugs? An analysis using human hepatocytes and liver S-9 fraction. Chem Res Toxicol. 2009;22(2):332–340.CrossRefGoogle ScholarPubMed
Zhao, SX, Dalvie, DK, Kelly, JM, et al. NADPH-dependent covalent binding of [3H]paroxetine to human liver microsomes and S-9 fractions: Identification of an electrophilic quinone metabolite of paroxetine. Chem Res Toxicol. 2007;20(11):1649–1657.CrossRefGoogle Scholar
Ring, BJ, Patterson, BE, Mitchell, MI, et al. Effect of tadalafil on cytochrome P450 3A4-mediated clearance: Studies in vitro and in vivo. Clin Pharmacol Ther. 2005;77(1):63–75.CrossRefGoogle ScholarPubMed
Erve, JC, Vashishtha, SC, DeMaio, W, et al. Metabolism of prazosin in rat, dog, and human liver microsomes and cryopreserved rat and human hepatocytes and characterization of metabolites by liquid chromatography/tandem mass spectrometry. Drug Metab Dispos. 2007;35(6):908–916.CrossRefGoogle ScholarPubMed
Hughes, AR, Brothers, CH, Mosteller, M, et al. Genetic association studies to detect adverse drug reactions: Abacavir hypersensitivity as an example. Pharmacogenomics. 2009;10(2):225–233.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×