1932

Abstract

Because of the inability to predict and quantify the risk of idiosyncratic adverse drug reactions (IADRs) and because reactive metabolites (RMs) are thought to be responsible for the pathogenesis of some IADRs, the potential for RM formation within new chemical entities is routinely examined with the ultimate goal of eliminating or reducing the liability through iterative design. Likewise, avoidance of structural alerts is almost a standard practice in drug design. However, the perceived safety concerns associated with the use of structural alerts and/or RM screening tools as standalone predictors of toxicity risks may be overexaggerated. Numerous marketed drugs form RMs but do not cause idiosyncratic toxicity. In this review article, we present a critique of the structural alert/RM concept as applied in drug discovery and evaluate the evidence linking structural alerts and RMs to observed toxic effects. Pragmatic risk mitigation strategies to aid the advancement of drug candidates that carry a RM liability are also discussed.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010814-124720
2015-01-06
2024-04-20
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/55/1/annurev-pharmtox-010814-124720.html?itemId=/content/journals/10.1146/annurev-pharmtox-010814-124720&mimeType=html&fmt=ahah

Literature Cited

  1. Orr ST, Ripp SL, Ballard TE, Henderson JL, Scott DO. 1.  et al. 2012. Mechanism-based inactivation (MBI) of cytochrome P450 enzymes: structure-activity relationships and discovery strategies to mitigate drug-drug interaction risks. J. Med. Chem. 55:4896–933 [Google Scholar]
  2. Dobo KL, Obach RS, Luffer-Atlas D, Bercu JP. 2.  2009. A strategy for the risk assessment of human genotoxic metabolites. Chem. Res. Toxicol. 22:348–56 [Google Scholar]
  3. Evans DC, Baillie TA. 3.  2005. Minimizing the potential for metabolic activation as an integral part of drug design. Curr. Opin. Drug Discov. Dev. 8:44–50 [Google Scholar]
  4. Guengerich FP, MacDonald JS. 4.  2007. Applying mechanisms of chemical toxicity to predict drug safety. Chem. Res. Toxicol. 20:344–69 [Google Scholar]
  5. Park BK, Boobis A, Clarke S, Goldring CE, Jones D. 5.  et al. 2011. Managing the challenge of chemically reactive metabolites in drug development. Nat. Rev. Drug Discov. 10:292–306 [Google Scholar]
  6. Stachulski AV, Baillie TA, Park BK, Obach RS, Dalvie DK. 6.  et al. 2013. The generation, detection, and effects of reactive drug metabolites. Med. Res. Rev. 33:985–1080 [Google Scholar]
  7. Lucas A, Nolan D, Mallal S. 7.  2007. HLA-B*5701 screening for susceptibility to abacavir hypersensitivity. J. Antimicrob. Chemother. 59:591–93 [Google Scholar]
  8. Singer JB, Lewitzky S, Leroy E, Yang F, Zhao X. 8.  et al. 2010. A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat. Genet. 42:711–14 [Google Scholar]
  9. Haas DW, Mootsikapun P, Ruxrungtham K, Podzamczer D. 9.  Nevirapine Toxicogenomics Study Team 2012. Clinical perspectives on human genetic screening to prevent nevirapine toxicity. Pers. Med. 9:773–82 [Google Scholar]
  10. Uetrecht J, Naisbitt DJ. 10.  2013. Idiosyncratic adverse drug reactions: current concepts. Pharmacol. Rev. 65:779–808 [Google Scholar]
  11. Park BK, Kitteringham NR, Maggs JL, Pirmohamed M, Williams DP. 11.  2005. The role of metabolic activation in drug-induced hepatotoxicity. Annu. Rev. Pharmacol. Toxicol. 45:177–202 [Google Scholar]
  12. Ng W, Lobach AR, Zhu X, Chen X, Liu F. 12.  et al. 2012. Animal models of idiosyncratic drug reactions. Adv. Pharmacol. 63:81–135 [Google Scholar]
  13. Evans DC, Watt AP, Nicoll-Griffith DA, Baillie TA. 13.  2004. Drug-protein adducts: an industry perspective on minimizing the potential for drug bioactivation in drug discovery and development. Chem. Res. Toxicol. 17:3–16 [Google Scholar]
  14. Argikar UA, Mangold JB, Harriman SP. 14.  2011. Strategies and chemical design approaches to reduce the potential for formation of reactive metabolic species. Curr. Top. Med. Chem. 11:419–49 [Google Scholar]
  15. Kalgutkar AS, Gardner I, Obach RS, Shaffer CL, Callegari E. 15.  et al. 2005. A comprehensive listing of bioactivation pathways of organic functional groups. Curr. Drug Metab. 6:161–225 [Google Scholar]
  16. Stepan AF, Walker DP, Bauman J, Price DA, Baillie TA. 16.  et al. 2011. 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. 24:1345–410 [Google Scholar]
  17. Shamovsky I, Ripa L, Blomberg N, Eriksson LA, Hansen P. 17.  et al. 2012. Theoretical studies of chemical reactivity of metabolically activated forms of aromatic amines toward DNA. Chem. Res. Toxicol. 25:2236–52 [Google Scholar]
  18. Liu ZC, Uetrecht JP. 18.  1995. Clozapine is oxidized by activated human neutrophils to a reactive nitrenium ion that irreversibly binds to the cells. J. Pharmacol. Exp. Ther. 275:1476–83 [Google Scholar]
  19. Gardner I, Leeder JS, Chin T, Zahid N, Uetrecht JP. 19.  1998. A comparison of the covalent binding of clozapine and olanzapine to human neutrophils in vitro and in vivo. Mol. Pharmacol. 53:999–1008 [Google Scholar]
  20. Uetrecht J, Zahid N, Tehim A, Fu JM, Rakhit S. 20.  1997. Structural features associated with reactive metabolite formation in clozapine analogues. Chem. Biol. Interact. 104:117–29 [Google Scholar]
  21. Kalgutkar AS, Vaz AD, Lame ME, Henne KR, Soglia J. 21.  et al. 2005. Bioactivation of the nontricyclic antidepressant nefazodone to a reactive quinone-imine species in human liver microsomes and recombinant cytochrome P450 3A4. Drug Metab. Dispos. 33:243–53 [Google Scholar]
  22. Damsten MC, de Vlieger JS, Niessen WM, Irth H, Vermeulen NP, Commandeur JN. 22.  2008. Trimethoprim: novel reactive intermediates and bioactivation pathways by cytochrome P450s. Chem. Res. Toxicol. 21:2181–87 [Google Scholar]
  23. Obach RS, Kalgutkar AS, Ryder TF, Walker GS. 23.  2008. In vitro metabolism and covalent binding of enol-carboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: insights into the hepatotoxicity of sudoxicam. Chem. Res. Toxicol. 21:1890–99 [Google Scholar]
  24. Davies NM, Skjodt NM. 24.  1999. Clinical pharmacokinetics of meloxicam: a cyclo-oxygenase-2 preferential nonsteroidal anti-inflammatory drug. Clin. Pharmacokinet. 36:115–26 [Google Scholar]
  25. Bell JA, Dallas FA, Jenner WN, Martin LE. 25.  1980. The metabolism of ranitidine in animals and man. Biochem. Soc. Trans. 8:93 [Google Scholar]
  26. Diao L, Shu Y, Polli JE. 26.  2010. Uptake of pramipexole by human organic cation transporters. Mol. Pharm. 7:1342–47 [Google Scholar]
  27. Dieckhaus CM, Thompson CD, Roller SG, Macdonald TL. 27.  2002. Mechanisms of idiosyncratic drug reactions: the case of felbamate. Chem. Biol. Interact. 142:99–117 [Google Scholar]
  28. Thompson CD, Barthen MT, Hopper DW, Miller TA, Quigg M. 28.  et al. 1999. Quantification in patient urine samples of felbamate and three metabolites: acid carbamate and two mercapturic acids. Epilepsia 40:769–76 [Google Scholar]
  29. Testa L, Bhindi R, Agostoni P, Abbate A, Zoccai GG, van Gaal WJ. 29.  2007. The direct thrombin inhibitor ximelagatran/melagatran: a systematic review on clinical applications and an evidence based assessment of risk benefit profile. Exp. Opin. Drug Saf. 6:397–406 [Google Scholar]
  30. McKenney JM, Proctor JD, Harris S, Chinchili VM. 30.  1994. A comparison of the efficacy and toxic effects of sustained- versus immediate-release niacin in hypercholesterolemic patients. JAMA 271:672–77 [Google Scholar]
  31. Pieper JA. 31.  2003. Overview of niacin formulations: differences in pharmacokinetics, efficacy, and safety. Am. J. Health Syst. Pharm. 60:S9–14 [Google Scholar]
  32. Roecklein BA, Sacks HJ, Mortko H, Stables J. 32.  2007. Fluorofelbamate. Neurotherapeutics 4:97–101 [Google Scholar]
  33. Hartz RA, Ahuja VT, Zhuo X, Mattson RJ, Denhart DJ. 33.  et al. 2009. A strategy to minimize reactive metabolite formation: discovery of (S)-4-(1-cyclopropyl-2-methoxyethyl)-6-[6-(difluoromethoxy)-2,5-dimethylpyridin-3-ylamino]-5-oxo-4,5-dihydropyrazine-2-carbonitrile as a potent, orally bioavailable corticotropin-releasing factor-1 receptor antagonist. J. Med. Chem. 52:7653–68 [Google Scholar]
  34. Subramanian R, Lee MR, Allen JG, Bourbeau MP, Fotsch C. 34.  et al. 2010. Cytochrome P450–mediated epoxidation of 2-aminothiazole-based AKT inhibitors: identification of novel GSH adducts and reduction of metabolic activation through structural changes guided by in silico and in vitro screening. Chem. Res. Toxicol. 23:653–63 [Google Scholar]
  35. Kalgutkar AS, Mascitti V, Sharma R, Walker GW, Ryder T. 35.  et al. 2011. Intrinsic electrophilicity of a 4-substituted-5-cyano-6-(2-methylpyridin-3-yloxy)pyrimidine derivative: structural characterization of glutathione conjugates in vitro. Chem. Res. Toxicol. 24:269–78 [Google Scholar]
  36. Kalgutkar AR, Ryder TF, Walker GS, Orr ST, Cabral S. 36.  et al. 2013. Reactive metabolite trapping studies on imidazo- and 2-methylimidazo[2,1-b]thiazole-based inverse agonists of the ghrelin receptor. Drug Metab. Dispos. 41:1375–88 [Google Scholar]
  37. Hagmann WK. 37.  2008. The discovery of taranabant, a selective cannabinoid-1 receptor inverse agonist for the treatment of obesity. Arch. Pharm. 341:405–11 [Google Scholar]
  38. Sarabu R, Bizzarro FT, Corbett WL, Dvorozniak MT, Geng W. 38.  et al. 2012. Discovery of piragliatin—first glucokinase activator studied in type 2 diabetic patients. J. Med. Chem. 55:7021–36 [Google Scholar]
  39. Lennernas H. 39.  2003. Clinical pharmacokinetics of atorvastatin. Clin. Pharmacokinet. 42:1141–60 [Google Scholar]
  40. Nakayama S, Atsumi R, Takakusa H, Kobayashi Y, Kurihara A. 40.  et al. 2009. A zone classification system for risk assessment of idiosyncratic drug toxicity using daily dose and covalent binding. Drug Metab. Dispos. 37:1970–77 [Google Scholar]
  41. Dansette PM, Libraire J, Bertho G, Mansuy D. 41.  2009. Metabolic oxidative cleavage of thioesters: evidence for the formation of sulfenic acid intermediates in the bioactivation of the antithrombotic prodrugs ticlopidine and clopidogrel. Chem. Res. Toxicol. 22:369–73 [Google Scholar]
  42. Savi P, Zachayus JL, Delesque-Touchard N, Labouret C, Hervé C. 42.  et al. 2006. The active metabolite of clopidogrel disrupts P2Y12 receptor oligomers and partitions them out of lipid rafts. Proc. Natl. Acad. Sci. USA 103:11069–74 [Google Scholar]
  43. Wu G, Vashishtha SC, Erve JC. 43.  2010. Characterization of glutathione conjugates of duloxetine by mass spectrometry and evaluation of in silico approaches to rationalize the site of conjugation for thiophene containing drugs. Chem. Res. Toxicol. 23:1393–404 [Google Scholar]
  44. Haddock RE, Johnson AM, Langley PF, Nelson DR, Pope JA. 44.  et al. 1989. Metabolic pathways of paroxetine in animals and man and the comparative pharmacological properties of the metabolites. Acta Psychiatr. Scand. Suppl. 350:24–26 [Google Scholar]
  45. Venkatakrishnan K, Obach RS. 45.  2005. In vitro–in vivo extrapolation of CYP2D6 inactivation by paroxetine: prediction of nonstationary pharmacokinetics and drug interaction magnitude. Drug Metab. Dispos. 33:845–52 [Google Scholar]
  46. Zhao SX, Dalvie DK, Kelly JM, Soglia JR, Frederick KS. 46.  et al. 2007. 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. 20:1649–57 [Google Scholar]
  47. Chen Q, Ngui JS, Doss GA, Wang RW, Cai X. 47.  et al. 2002. Cytochrome P450 3A4–mediated bioactivation of raloxifene: irreversible enzyme inhibition and thiol adduct formation. Chem. Res. Toxicol. 15:907–14 [Google Scholar]
  48. Hucker HB, Stauffer SC, Balletto AJ, White SD, Zacchei AG, Arison BH. 48.  1978. Physiological disposition and metabolism of cyclobenzaprine in the rat, dog, rhesus monkey, and man. Drug Metab. Dispos. 6:659–72 [Google Scholar]
  49. Devineni D, Curtin CR, Polidori D, Gutierrez MJ, Murphy J. 49.  et al. 2013. Pharmacokinetics and pharmacodynamics of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in subjects with type 2 diabetes mellitus. J. Clin. Pharmacol. 53:601–10 [Google Scholar]
  50. Zhang D, He K, Raghavan N, Wang L, Mitroka J. 50.  et al. 2009. Comparative metabolism of 14C-labeled apixaban in mice, rats, rabbits, dogs and humans. Drug Metab. Dispos. 37:1738–48 [Google Scholar]
  51. Weinz C, Schwarz T, Kubiza D, Mueck W, Lang D. 51.  2009. Metabolism and excretion of rivaroxaban, an oral, direct factor Xa inhibitor, in rats, dogs, and humans. Drug Metab. Dispos. 37:1056–64 [Google Scholar]
  52. Stangier J. 52.  2008. Clinical pharmacokinetics and pharmacodynamics of the oral direct thrombin inhibitor dabigatran etexilate. Clin. Pharmacokinet. 47:285–95 [Google Scholar]
  53. 53. Xtandi® [package insert] Northbrook, IL: Astellas Pharma Inc. 2012.
  54. Hoffmann M, Kasserra C, Reyes J, Schafer P, Kosek J. 54.  et al. 2013. Absorption, metabolism and excretion of [14C]pomalidomide in humans following oral administration. Cancer Chemother. Pharmacol. 71:489–501 [Google Scholar]
  55. Chowdhury G, Shibata N, Yamazaki H, Guengerich FP. 55.  2014. Human cytochrome P450 oxidation of 5-hydroxythalidomide and pomalidomide, an amino analogue of thalidomide. Chem. Res. Toxicol. 27:147–56 [Google Scholar]
  56. 56. Cometriq® [package insert] South San Francisco, CA: Exelixis Inc. 2012.
  57. 57. Votrient® [package insert] Research Triangle Park, NC: GlaxoSmithKline 2013.
  58. 58. Iclusig® [package insert] Cambridge, MA: Ariad Pharmaceuticals Inc. 2012.
  59. Kenny JR, Mukadam S, Zhang C, Tay S, Collins C. 59.  et al. 2012. Drug-drug interaction potential of marketed oncology drugs: in vitro assessment of time-dependent cytochrome P450 inhibition, reactive metabolite formation and drug-drug interaction. Pharm. Res. 29:1960–76 [Google Scholar]
  60. Narasimhan NI, Dorer DJ, Niland K, Haluska F, Sonnichsen D. 60.  2013. Effects of ketoconazole on the pharmacokinetics of ponatinib in healthy subjects. J. Clin. Pharmacol. 53:974–81 [Google Scholar]
  61. Martin P, Oliver S, Kennedy SJ, Partridge E, Hutchison M. 61.  et al. 2012. Pharmacokinetics of vandetanib: three phase I studies in healthy subjects. Clin. Ther. 34:221–37 [Google Scholar]
  62. Zhou HJ, Aujay MA, Bennett MK, Dajee M, Demo SD. 62.  et al. 2009. Design and synthesis of an orally bioavailable and selective peptide epoxyketone proteasome inhibitor (PR-047). J. Med. Chem. 52:3028–38 [Google Scholar]
  63. Wang Z, Yang J, Kirk C, Fang Y, Alsina M. 63.  et al. 2013. Clinical pharmacokinetics, metabolism, and drug-drug interaction of carfilzomib. Drug Metab. Dispos. 41:230–37 [Google Scholar]
  64. Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X. 64.  et al. 2011. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 117:6287–96 [Google Scholar]
  65. Dalvie D, Kang P, Zientek M, Xiang C, Zhou S, Obach RS. 65.  2008. Effect of intestinal glucuronidation in limiting hepatic exposure and bioactivation of raloxifene in humans and rats. Chem. Res. Toxicol. 21:2260–71 [Google Scholar]
  66. Obach RS, Kalgutkar AS, Soglia JR, Zhao SX. 66.  2008. 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. 21:1814–22 [Google Scholar]
  67. Dalvie D, Obach RS, Kang P, Prakash C, Loi CM. 67.  et al. 2009. Assessment of three human in vitro systems in the generation of major human excretory and circulating metabolites. Chem. Res. Toxicol. 22:357–68 [Google Scholar]
  68. Genève J, Degott C, Letteron P, Tinel M, Descatoire V. 68.  et al. 1987. Metabolic activation of the tricyclic antidepressant amineptine: II. Protective role of glutathione against in vitro and in vivo covalent binding. Biochem. Pharmacol. 36:331–37 [Google Scholar]
  69. Fromenty B, Pessayre D. 69.  1995. Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. Pharmacol. Ther. 67:101–54 [Google Scholar]
  70. Farid NA, Kurihara A, Wrighton SA. 70.  2010. Metabolism and disposition of the thienopyridine antiplatelet drugs ticlopidine, clopidogrel, and prasugrel in humans. J. Clin. Pharmacol. 50:126–42 [Google Scholar]
  71. 71. Plavix® [package insert] Bridgewater, NJ: Sanofi-Aventis 2013.
  72. 72. Mekinist [package insert] Research Triangle Park, NC: GlaxoSmithKline 2014.
  73. Bauman JS, Kelly JM, Tripathy S, Zhao SX, Lam WM. 73.  et al. 2009. 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. 22:332–40 [Google Scholar]
  74. Thompson RA, Isin EM, Li Y, Weidolf L, Page K. 74.  et al. 2012. In vitro approach to assess the potential for risk of idiosyncratic adverse reactions caused by candidate drugs. Chem. Res. Toxicol. 25:1616–32 [Google Scholar]
  75. Teng WC, Oh JW, New LS, Wahlin MD, Nelson SD. 75.  et al. 2010. Mechanism-based inactivation of cytochrome P450 3A4 by lapatinib. Mol. Pharmacol. 78:693–703 [Google Scholar]
  76. Didiuk MT, Griffith DA, Benbow JW, Liu KK, Walker DP. 76.  et al. 2009. Short-acting 5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one derivatives as orally-active calcium-sensing receptor antagonists. Bioorg. Med. Chem. Lett. 19:4555–59 [Google Scholar]
  77. Kalgutkar AS, Griffith DA, Ryder T, Sun H, Miao Z. 77.  et al. 2010. Discovery tactics to mitigate toxicity risks due to reactive metabolite formation with 2-(2-hydroxyaryl)-5-(trifluoromethyl)pyrido[4,3-d]pyrimidin-4(3H)-one derivatives, potent calcium-sensing receptor antagonists and clinical candidate(s) for the treatment of osteoporosis. Chem. Res. Toxicol. 23:1115–26 [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010814-124720
Loading
/content/journals/10.1146/annurev-pharmtox-010814-124720
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error