Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Managing the challenge of chemically reactive metabolites in drug development

Key Points

  • Metabolism of drugs can generate metabolites that are chemically reactive towards cellular molecules and have the potential to alter biological function and initiate serious adverse drug reactions.

  • This Review details methods for the detection of chemically reactive metabolites (CRMs), and outlines the current industrial and academic knowledge about structural alerts.

  • The physiological response to bioactivation is discussed in the context of the toxicological response and how hypersensitivity reactions may occur.

  • We also discuss the management of CRMs during drug development, taking into account whether the currently used CRM decision trees are relevant to the challenges posed in drug development.

  • Earlier iteration between medicinal chemistry and drug metabolism can eliminate perceived reactive metabolite-mediated chemical liabilities without comprising pharmacological activity or the need for extensive safety evaluation beyond standard practices.

  • Finally, we address CRM-related decision-making based on minimal data (avoidance strategy) and how to make decisions based on covalent binding and other data. The implications for drug regulation are outlined.

Abstract

The normal metabolism of drugs can generate metabolites that have intrinsic chemical reactivity towards cellular molecules, and therefore have the potential to alter biological function and initiate serious adverse drug reactions. Here, we present an assessment of the current approaches used for the evaluation of chemically reactive metabolites. We also describe how these approaches are being used within the pharmaceutical industry to assess and minimize the potential of drug candidates to cause toxicity. At early stages of drug discovery, iteration between medicinal chemistry and drug metabolism can eliminate perceived reactive metabolite-mediated chemical liabilities without compromising pharmacological activity or the need for extensive safety evaluation beyond standard practices. In the future, reactive metabolite evaluation may also be useful during clinical development for improving clinical risk assessment and risk management. Currently, there remains a huge gap in our understanding of the basic mechanisms that underlie chemical stress-mediated adverse reactions in humans. This Review summarizes our views on this complex topic, and includes insights into practices considered by the pharmaceutical industry.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: An ideal working relationship between chemistry and drug metabolism.
Figure 2: Examples of possible decision scheme structures for handling bioactivation information in various stages of drug research.

Similar content being viewed by others

References

  1. Lasser, K. E. et al. Timing of new black box warnings and withdrawals for prescription medications. JAMA 287, 2215–2220 (2002).

    Article  PubMed  Google Scholar 

  2. Ostapowicz, G. et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann. Intern. Med. 137, 947–954 (2002).

    Article  PubMed  Google Scholar 

  3. Adams, D. H., Ju, C., Ramaiah, S. K., Uetrecht, J. & Jaeschke, H. Mechanisms of immune-mediated liver injury. Toxicol. Sci. 115, 307–321 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Li, J. & Uetrecht, J. P. The danger hypothesis applied to idiosyncratic drug reactions. Handb. Exp. Pharmacol. 196, 493–509 (2010).

    Article  CAS  Google Scholar 

  5. Park, B. K., Kitteringham, N. R., Maggs, J. L., Pirmohamed, M. & Williams, D. P. The role of metabolic activation in drug-induced hepatotoxicity. Annu. Rev. Pharmacol. Toxicol. 45, 177–202 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Erve, J. C. Chemical toxicology: reactive intermediates and their role in pharmacology and toxicology. Expert Opin. Drug Metab. Toxicol. 2, 923–946 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Guengerich, F. P. & MacDonald, J. S. Applying mechanisms of chemical toxicity to predict drug safety. Chem. Res. Toxicol. 20, 344–369 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Kalgutkar, A. S. & Soglia, J. R. Minimising the potential for metabolic activation in drug discovery. Expert Opin. Drug Metab. Toxicol. 1, 91–142 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Nelson, S. D. Structure toxicity relationships — how useful are they in predicting toxicities of new drugs? Adv. Exp. Med. Biol. 500, 33–43 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Walgren, J. L., Mitchell, M. D. & Thompson, D. C. Role of metabolism in drug-induced idiosyncratic hepatotoxicity. Crit. Rev. Toxicol. 35, 325–361 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Tirmenstein, M. A. & Nelson, S. D. Subcellular binding and effects on calcium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 3′-hydroxyacetanilide, in mouse liver. J. Biol. Chem. 264, 9814–9819 (1989).

    CAS  PubMed  Google Scholar 

  12. Liebler, D. C. Protein damage by reactive electrophiles: targets and consequences. Chem. Res. Toxicol. 21, 117–128 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Kenna, J. G. in Drug Induced Liver Disease (eds Kaplowitz, N., & DeLeve, L. D.) 465–484 (Marcel Dekker, New York, 2007).

    Book  Google Scholar 

  14. Obach, R. S., Kalgutkar, A. S., Soglia, J. R. & Zhao, S. X. 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. 21, 1814–1822 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Coles, B. et al. The spontaneous and enzymatic reaction of N-acetyl-p-benzoquinonimine with glutathione: a stopped-flow kinetic study. Arch. Biochem. Biophys. 264, 253–260 (1988).

    Article  CAS  PubMed  Google Scholar 

  16. Tingle, M. D., Mahmud, R., Maggs, J. L., Pirmohamed, M. & Park, B. K. Comparison of the metabolism and toxicity of dapsone in rat, mouse and man. J. Pharmacol. Exp. Ther. 283, 817–823 (1997).

    CAS  PubMed  Google Scholar 

  17. Nakayama, S. et al. A zone classification system for risk assessment of idiosyncratic drug toxicity using daily dose and covalent binding. Drug Metab. Dispos. 37, 1970–1977 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Usui, T., Mise, M., Hashizume, T., Yabuki, M. & Komuro, S. Evaluation of the potential for drug-induced liver injury based on in vitro covalent binding to human liver proteins. Drug Metab. Dispos. 37, 2383–2392 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. de Rooij, B. M., Boogaard, P. J., Commandeur, J. N., van Sittert, N. J. & Vermeulen, N. P. Allylmercapturic acid as urinary biomarker of human exposure to allyl chloride. Occup. Environ. Med. 54, 653–661 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nelson, E. B. The pharmacology and toxicology of meta-substituted acetanilide I: acute toxicity of 3-hydroxyacetanilide in mice. Res. Commun. Chem. Pathol. Pharmacol. 28, 447–456 (1980).

    CAS  PubMed  Google Scholar 

  21. Bauman, J. N. 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. 22, 332–340 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Mallal, S. et al. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 359, 727–732 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Chessman, D. et al. Human leukocyte antigen class I-restricted activation of CD8+ T cells provides the immunogenetic basis of a systemic drug hypersensitivity. Immunity 28, 822–832 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Evans, D. C., Watt, A. P., Nicoll-Griffith, D. A. & Baillie, T. A. Drug–protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development. Chem. Res. Toxicol. 17, 3–16 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Smith, D. A. & Obach, R. S. Metabolites in safety testing (MIST): considerations of mechanisms of toxicity with dose, abundance, and duration of treatment. Chem. Res. Toxicol. 22, 267–279 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Smith, D. A., Obach, R. S., Williams, D. P. & Park, B. K. Clearing the MIST (metabolites in safety testing) of time: the impact of duration of administration on drug metabolite toxicity. Chem. Biol. Interact. 179, 60–67 (2009).

    Article  CAS  PubMed  Google Scholar 

  27. Kalgutkar, A. S. et al. A comprehensive listing of bioactivation pathways of organic functional groups. Curr. Drug Metab. 6, 161–225 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Uetrecht, J. Prediction of a new drug's potential to cause idiosyncratic reactions. Curr. Opin. Drug Discov. Devel. 4, 55–59 (2001).

    CAS  PubMed  Google Scholar 

  29. Day, S. H. et al. A semi-automated method for measuring the potential for protein covalent binding in drug discovery. J. Pharmacol. Toxicol. Methods 52, 278–285 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Baillie, T. A. Metabolism and toxicity of drugs. Two decades of progress in industrial drug metabolism. Chem. Res. Toxicol. 21, 129–137 (2008).

    Article  PubMed  Google Scholar 

  31. Bateman, K. P. et al. MSE with mass defect filtering for in vitro and in vivo metabolite identification. Rapid Commun. Mass Spectrom. 21, 1485–1496 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Fontana, E., Dansette, P. M. & Poli, S. M. Cytochrome p450 enzymes mechanism based inhibitors: common sub-structures and reactivity. Curr. Drug Metab. 6, 413–454 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Kalgutkar, A. S., Obach, R. S. & Maurer, T. S. 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, 407–447 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Danciu, T. E. & Whitman, M. Oxidative stress drives disulfide bond formation between basic helix–loop–helix transcription factors. J. Cell Biochem. 109, 417–424 (2010).

    CAS  PubMed  Google Scholar 

  35. Jacobs, A. T. & Marnett, L. J. Heat shock factor 1 attenuates 4-hydroxynonenal-mediated apoptosis: critical role for heat shock protein 70 induction and stabilization of Bcl-XL. J. Biol. Chem. 282, 33412–33420 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Lieu, H. T. et al. Reg2 inactivation increases sensitivity to Fas hepatotoxicity and delays liver regeneration post-hepatectomy in mice. Hepatology 44, 1452–1464 (2006).

    Article  CAS  PubMed  Google Scholar 

  37. Bateman, K. P. et al. Detection of covalent adducts to cytochrome P450 3A4 using liquid chromatography mass spectrometry. Chem. Res. Toxicol. 17, 1356–1361 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Fang, J., Koen, Y. M. & Hanzlik, R. P. Bioinformatic analysis of xenobiotic reactive metabolite target proteins and their interacting partners. BMC Chem. Biol. 9, 5 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hanzlik, R. P., Koen, Y. M., Theertham, B., Dong, Y. & Fang, J. The reactive metabolite target protein database (TPDB) — a web-accessible resource. BMC Bioinformatics 8, 95 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gan, J., Harper, T. W., Hsueh, M. M., Qu, Q. & Humphreys, W. G. Dansyl glutathione as a trapping agent for the quantitative estimation and identification of reactive metabolites. Chem. Res. Toxicol. 18, 896–903 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Yan, Z., Maher, N., Torres, R. & Huebert, N. Use of a trapping agent for simultaneous capturing and high-throughput screening of both “soft” and “hard” reactive metabolites. Anal. Chem. 79, 4206–4214 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Ma, L., Wen, B., Ruan, Q. & Zhu, M. Rapid screening of glutathione-trapped reactive metabolites by linear ion trap mass spectrometry with isotope pattern-dependent scanning and postacquisition data mining. Chem. Res. Toxicol. 21, 1477–1483 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Ma, X. & Chan, E. C. Fluorescence-based liver microsomal assay for screening of pharmaceutical reactive metabolites using a glutathione conjugated 96-well plate. Bioconjug. Chem. 21, 46–55 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Fontana, R. J. et al. Acute liver failure associated with prolonged use of bromfenac leading to liver transplantation. The Acute Liver Failure Study Group. Liver Transpl. Surg. 5, 480–484 (1999).

    Article  CAS  PubMed  Google Scholar 

  45. Hunter, E. B., Johnston, P. E., Tanner, G., Pinson, C. W. & Awad, J. A. Bromfenac (Duract)-associated hepatic failure requiring liver transplantation. Am. J. Gastroenterol. 94, 2299–2301 (1999).

    Article  CAS  PubMed  Google Scholar 

  46. Moses, P. L. et al. Severe hepatotoxicity associated with bromfenac sodium. Am. J. Gastroenterol. 94, 1393–1396 (1999).

    Article  CAS  PubMed  Google Scholar 

  47. 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. 15, 213–220 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Miyanaga, M. et al. Effect of bromfenac ophthalmic solution on ocular inflammation following cataract surgery. Acta Ophthalmol. 87, 300–305 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Bakke, O. M., Manocchia, M., de Abajo, F., Kaitin, K. I. & Lasagna, L. Drug safety discontinuations in the United Kingdom, the United States, and Spain from 1974 through 1993: a regulatory perspective. Clin. Pharmacol. Ther. 58, 108–117 (1995).

    Article  CAS  PubMed  Google Scholar 

  50. Berson, A. et al. Inhibition by nilutamide of the mitochondrial respiratory chain and ATP formation. Possible contribution to the adverse effects of this antiandrogen. J. Pharmacol. Exp. Ther. 270, 167–176 (1994).

    CAS  PubMed  Google Scholar 

  51. Wen, B. et al. Comparison of in vitro bioactivation of flutamide and its cyano analogue: evidence for reductive activation by human NADPH: cytochrome P450 reductase. Chem. Res. Toxicol. 21, 2393–2406 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Schellhammer, P. F. et al. Clinical benefits of bicalutamide compared with flutamide in combined androgen blockade for patients with advanced prostatic carcinoma: final report of a double-blind, randomized, multicenter trial. Casodex Combination Study Group. Urology 50, 330–336 (1997).

    Article  CAS  PubMed  Google Scholar 

  53. Coe, K. J. et al. Comparison of the cytotoxicity of the nitroaromatic drug flutamide to its cyano analogue in the hepatocyte cell line TAMH: evidence for complex I inhibition and mitochondrial dysfunction using toxicogenomic screening. Chem. Res. Toxicol. 20, 1277–1290 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bailey, M. J. & Dickinson, R. G. Acyl glucuronide reactivity in perspective: biological consequences. Chem. Biol. Interact. 145, 117–137 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Boelsterli, U. A. Nimesulide and hepatic adverse effects: roles of reactive metabolites and host factors. Int. J. Clin. Pract. 128, 30–36 (2002).

    CAS  Google Scholar 

  56. Boelsterli, U. A. Mechanisms of NSAID-induced hepatotoxicity: focus on nimesulide. Drug Saf. 25, 633–648 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Skonberg, C., Olsen, J., Madsen, K. G., Hansen, S. H. & Grillo, M. P. Metabolic activation of carboxylic acids. Expert Opin. Drug Metab. Toxicol. 4, 425–438 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Boelsterli, U. A. Xenobiotic acyl glucuronides and acyl CoA thioesters as protein-reactive metabolites with the potential to cause idiosyncratic drug reactions. Curr. Drug Metab. 3, 439–450 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Kitteringham, N. R. et al. Hepatocellular response to chemical stress in CD-1 mice: induction of early genes and γ-glutamylcysteine synthetase. Hepatology 32, 321–333 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Dambach, D. M., Durham, S. K., Laskin, J. D. & Laskin, D. L. Distinct roles of NF-κB p50 in the regulation of acetaminophen-induced inflammatory mediator production and hepatotoxicity. Toxicol. Appl. Pharmacol. 211, 157–165 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Xu, Z. et al. Liver-specific inactivation of the Nrf1 gene in adult mouse leads to nonalcoholic steatohepatitis and hepatic neoplasia. Proc. Natl Acad. Sci. USA 102, 4120–4125 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Copple, I. M., Goldring, C. E., Kitteringham, N. R. & Park, B. K. The keap1-nrf2 cellular defense pathway: mechanisms of regulation and role in protection against drug-induced toxicity. Handb. Exp. Pharmacol. 196, 233–266 (2010).

    Article  CAS  Google Scholar 

  63. Chevillard, G., Nouhi, Z., Anna, D., Paquet, M. & Blank, V. Nrf3-deficient mice are not protected against acute lung and adipose tissue damages induced by butylated hydroxytoluene. FEBS Lett. 584, 923–928.

  64. Cheng, J., Ma, X., Krausz, K. W., Idle, J. R. & Gonzalez, F. J. Rifampicin-activated human pregnane X receptor and CYP3A4 induction enhance acetaminophen-induced toxicity. Drug Metab. Dispos. 37, 1611–1621 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Grosch, S., Fritz, G. & Kaina, B. Apurinic endonuclease (Ref-1) is induced in mammalian cells by oxidative stress and involved in clastogenic adaptation. Cancer Res. 58, 4410–4416 (1998).

    CAS  PubMed  Google Scholar 

  66. Bykov, V. J., Lambert, J. M., Hainaut, P. & Wiman, K. G. Mutant p53 rescue and modulation of p53 redox state. Cell Cycle 8, 2509–2517 (2009).

    Article  CAS  PubMed  Google Scholar 

  67. Rzymski, T., Milani, M., Singleton, D. C. & Harris, A. L. Role of ATF4 in regulation of autophagy and resistance to drugs and hypoxia. Cell Cycle 8, 3838–3847 (2009).

    Article  CAS  PubMed  Google Scholar 

  68. Pessler-Cohen, D. et al. GLUT4 repression in response to oxidative stress is associated with reciprocal alterations in C/EBP α and δ isoforms in 3T3-L1 adipocytes. Arch. Physiol. Biochem. 112, 3–12 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Stamper, B. D., Bammler, T. K., Beyer, R. P., Farin, F. M. & Nelson, S. D. Differential regulation of mitogen-activated protein kinase pathways by acetaminophen and its nonhepatotoxic regioisomer 3′-hydroxyacetanilide in TAMH cells. Toxicol. Sci. 116, 164–173 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. McCubrey, J. A., Lahair, M. M. & Franklin, R. A. Reactive oxygen species-induced activation of the MAP kinase signaling pathways. Antioxid. Redox Signal. 8, 1775–1789 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Copple, I. M. et al. The hepatotoxic metabolite of acetaminophen directly activates the Keap1-Nrf2 cell defense system. Hepatology 48, 1292–1301 (2008).

    Article  CAS  PubMed  Google Scholar 

  72. Dinkova-Kostova, A. T. et al. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proc. Natl Acad. Sci. USA 99, 11908–11913 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hong, F., Sekhar, K. R., Freeman, M. L. & Liebler, D. C. Specific patterns of electrophile adduction trigger Keap1 ubiquitination and Nrf2 activation. J. Biol. Chem. 280, 31768–31775 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Itoh, K., Mimura, J. & Yamamoto, M. Discovery of the negative regulator of Nrf2, Keap1: A historical overview. Antioxid. Redox Signal. 13, 1665–1678 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Goldring, C. E. et al. Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice. Hepatology 39, 1267–1276 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Nassif, A. et al. Toxic epidermal necrolysis: effector cells are drug-specific cytotoxic T cells. J. Allergy Clin. Immunol. 114, 1209–1215 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Farrell, J. et al. Characterization of sulfamethoxazole and sulfamethoxazole metabolite-specific T-cell responses in animals and humans. J. Pharmacol. Exp. Ther. 306, 229–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  78. Schnyder, B. et al. Recognition of sulfamethoxazole and its reactive metabolites by drug-specific CD4+ T cells from allergic individuals. J. Immunol. 164, 6647–6654 (2000).

    Article  CAS  PubMed  Google Scholar 

  79. Sanderson, J. P. et al. Sulfamethoxazole and its metabolite nitroso sulfamethoxazole stimulate dendritic cell costimulatory signaling. J. Immunol. 178, 5533–5542 (2007).

    Article  CAS  PubMed  Google Scholar 

  80. Lavergne, S. N. et al. Drug metabolite-specific lymphocyte responses in sulfamethoxazole allergic patients with cystic fibrosis. Chem. Res. Toxicol. 23, 1009–1011 (2010).

    Article  CAS  PubMed  Google Scholar 

  81. Park, B. K., Pirmohamed, M. & Kitteringham, N. R. Role of drug disposition in drug hypersensitivity: a chemical, molecular, and clinical perspective. Chem. Res. Toxicol. 11, 969–988 (1998).

    Article  CAS  PubMed  Google Scholar 

  82. Kitteringham, N. R., Kenna, J. G. & Park, B. K. Detection of autoantibodies directed against human hepatic endoplasmic-reticulum in sera from patients with halothane-associated hepatitis. Br. J. Clin. Pharmacol. 40, 379–386 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Cheng, L., You, Q., Yin, H., Holt, M. P. & Ju, C. Involvement of natural killer T cells in halothane-induced liver injury in mice. Biochem. Pharmacol. 80, 255–261 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. You, Q., Cheng, L. & Ju, C. Generation of T cell responses targeting the reactive metabolite of halothane in mice. Toxicol. Lett. 194, 79–85 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Maria, V. A. & Victorino, R. M. Immunological investigation in hepatic drug reactions. Clin. Exp. Allergy 28 (Suppl. 4), 71–77 (1998).

    CAS  PubMed  Google Scholar 

  86. Aithal, G. P. et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology 39, 1430–1440 (2004).

    Article  CAS  PubMed  Google Scholar 

  87. Daly, A. K. et al. Genetic susceptibility to diclofenac-induced hepatotoxicity: contribution of UGT2B7, CYP2C8, and ABCC2 genotypes. Gastroenterology 132, 272–281 (2007).

    Article  CAS  PubMed  Google Scholar 

  88. Hanzlik, R. P., Fang, J. & Koen, Y. M. Filling and mining the reactive metabolite target protein database. Chem. Biol. Interact. 179, 38–44 (2009).

    Article  CAS  PubMed  Google Scholar 

  89. Nakamura, Y. et al. Redox regulation of glutathione S-transferase induction by benzyl isothiocyanate: correlation of enzyme induction with the formation of reactive oxygen intermediates. Cancer Res. 60, 219–225 (2000).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the Medical Research Council (MRC) and The Association of British Pharmaceutical Companies (ABPI). The Merck retrospective analysis was conducted in-house with a crossfunctional team led by D.N.-G. with recognition to J. Monroe and F. Sistare for curating the safety assessment data, members of DMPK for CB data and the sponsorship of L. Shipley, J. Harrelson and D. Dean. An industrial–academic workshop that was held in Liverpool, UK, on 13 and 14 April 2010 formed the initiative for the development of this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to B. Kevin Park or Donald J. Tweedie.

Ethics declarations

Competing interests

Scott Obach is an employee and shareholder of Pfizer — a company that is engaged in the discovery, development and sale of pharmaceuticals for profit.

Philip Routledge is head of the section of Pharmacology, Therapeutics & Toxicology at Cardiff University, UK, which has a clinical lectureship that is unconditionally part-funded by AstraZeneca under the Association for British Pharmaceutical Industries (ABPI) Clinical Pharmacology Training Scheme and part-funded by the Welsh Assembly Government.

Deborah Nicoll-Griffith is an employee of Merck & Co.

All other authors declare no competing financial interests.

Related links

Related links

DATABASES

Target Protein Database

Rights and permissions

Reprints and permissions

About this article

Cite this article

Park, B., Boobis, A., Clarke, S. et al. Managing the challenge of chemically reactive metabolites in drug development. Nat Rev Drug Discov 10, 292–306 (2011). https://doi.org/10.1038/nrd3408

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3408

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research