1932

Abstract

Drug-induced liver injury (DILI) is a leading cause of attrition during the early and late stages of drug development and after a drug is marketed. DILI is generally classified as either intrinsic or idiosyncratic. Intrinsic DILI is dose dependent and predictable (e.g., acetaminophen toxicity). However, predicting the occurrence of idiosyncratic DILI, which has a very low incidence and is associated with severe liver damage, is difficult because of its complex nature and the poor understanding of its mechanism. Considering drug metabolism and pharmacokinetics, we established experimental animal models of DILI for 14 clinical drugs that cause idiosyncratic DILI in humans, which is characterized by the formation of reactive metabolites and the involvement of both innate and adaptive immunity. On the basis of the biomarker data obtained from the animal models, we developed a cell-based assay system that predicts the potential risks of drugs for inducing DILI. These findings increase our understanding of the mechanisms of DILI and may help predict and prevent idiosyncratic DILI due to certain drugs.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-030220-015007
2021-01-06
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/61/1/annurev-pharmtox-030220-015007.html?itemId=/content/journals/10.1146/annurev-pharmtox-030220-015007&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Arrowsmith J. 2011. Trial watch: phase II failures: 2008–2010. Nat. Rev. Drug Discov. 10:328–29
    [Google Scholar]
  2. 2. 
    Arrowsmith J, Miller P. 2013. Phase II and phase III attrition rates 2011–2012. Nat. Rev. Drug Discov. 12:569
    [Google Scholar]
  3. 3. 
    Cook D, Brown D, Alexander R, March R, Morgan P et al. 2014. Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework. Nat. Rev. Drug Discov. 13:419–31
    [Google Scholar]
  4. 4. 
    Siramshetty VB, Nickel J, Omieczynski C, Gohlke B-O, Drwal MN et al. 2016. WITHDRAWN—a resource for withdrawn and discontinued drugs. Nucl. Acids Res. 44:D1080–86
    [Google Scholar]
  5. 5. 
    Lee WM. 2003. Drug-induced hepatotoxicity. N. Engl. J. Med. 349:474–85
    [Google Scholar]
  6. 6. 
    Park BK, Kitteringham NR, Maggs JL, Pirmohamed M, Williams DP 2005. The role of metabolic activation in drug-induced hepatotoxicity. Annu. Rev. Pharmacol. Toxicol. 45:177–202
    [Google Scholar]
  7. 7. 
    Kaplowitz N. 2005. Idiosyncratic drug hepatotoxicity. Nat. Rev. Drug Discov. 4:489–99
    [Google Scholar]
  8. 8. 
    Giri S, Nieber K, Bader A 2010. Hepatotoxicity and hepatic metabolism of available drugs: current problems and possible solutions in preclinical stages. Expert Opin. Drug Metab. Toxicol. 6:895–917
    [Google Scholar]
  9. 9. 
    Björnsson E, Olsson R. 2005. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology 42:481–89
    [Google Scholar]
  10. 10. 
    Kaku K, Enya K, Nakaya R, Ohira T, Matsuno R 2015. Efficacy and safety of fasiglifam (TAK-875), a G protein-coupled receptor 40 agonist, in Japanese patients with type 2 diabetes inadequately controlled by diet and exercise: a randomized, double-blind, placebo-controlled, phase III trial. Diabetes Obes. Metabol. 17:675–81
    [Google Scholar]
  11. 11. 
    Li AP. 2002. A review of the common properties of drugs with idiosyncratic hepatotoxicity and the “multiple determinant hypothesis” for the manifestation of idiosyncratic drug toxicity. Chem. Biol. Interact. 142:7–23
    [Google Scholar]
  12. 12. 
    Zhang J, Doshi U, Suzuki A, Chang C-W, Borlak J et al. 2016. Evaluation of multiple mechanism-based toxicity endpoints in primary cultured human hepatocytes for the identification of drugs with clinical hepatotoxicity: results from 152 marketed drugs with known liver injury profiles. Chem. Biol. Interact. 255:3–11
    [Google Scholar]
  13. 13. 
    Kobayashi E, Kobayashi M, Tsuneyama K, Fukami T, Nakajima M, Yokoi T 2009. Halothane-induced liver injury is mediated by interleukin-17 in mice. Toxicol. Sci. 111:302–10
    [Google Scholar]
  14. 14. 
    Higuchi S, Yano A, Takai S, Tsuneyama K, Fukami T et al. 2012. Metabolic activation and inflammation reactions involved in carbamazepine-induced liver injury. Toxicol. Sci. 130:4–16
    [Google Scholar]
  15. 15. 
    Sasaki E, Matsuo K, Iida A, Tsuneyama K, Fukami T et al. 2013. A novel mouse model for phenytoin-induced liver injury: involvement of immune-related factors and P450-mediated metabolism. Toxicol. Sci. 136:250–63
    [Google Scholar]
  16. 16. 
    Chen M, Vijay V, Shi Q, Liu Z, Fang H, Tong W 2011. FDA-approved drug labeling for the study of drug-induced liver injury. Drug Discov. Today 16:697–703
    [Google Scholar]
  17. 17. 
    Oda S, Matsuo K, Nakajima A, Yokoi T 2016. A novel cell-based assay for evaluation of immune- and inflammatory-related gene expression as biomarkers for the risk assessment of drug-induced liver injury. Toxicol. Lett. 241:60–70
    [Google Scholar]
  18. 18. 
    Luo G, Shen Y, Yang L, Lu A, Xiang Z 2017. A review of drug-induced liver injury databases. Arch. Toxicol. 91:3039–49
    [Google Scholar]
  19. 19. 
    Chen M, Suzuki A, Thakkar S, Yu K, Hu C, Tong W 2016. DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans. Drug Discov. Today 21:648–53
    [Google Scholar]
  20. 20. 
    Uetrecht J. 2003. Bioactivation. Cytochrome P450 and Other Enzymes in Drug Discovery and Development J Lee, RS Obach, MB Fisher 87–145 Boca Raton, FL: CRC Press
    [Google Scholar]
  21. 21. 
    You Q, Cheng L, Reilly TP, Wegmann D, Ju C 2006. Role of neutrophils in a mouse model of halothane-induced liver injury. Hepatology 44:1421–31
    [Google Scholar]
  22. 22. 
    Proctor WR, Chakraborty M, Chea LS, Morrison JC, Berkson JD et al. 2013. Eosinophils mediate the pathogenesis of halothane-induced liver injury in mice. Hepatology 57:2026–36
    [Google Scholar]
  23. 23. 
    Proctor WR, Chakraborty M, Fullerton AM, Korrapati MC, Ryan PM et al. 2016. Thymic stromal lymphopoietin and interleukin-4 mediate the pathogenesis of halothane-induced liver injury in mice. Hepatology 60:1741–52
    [Google Scholar]
  24. 24. 
    Kurth MJ, Yokoi T, Gershwin ME 2014. Halothane-induced hepatitis: paradigm or paradox for drug-induced liver injury. Hepatology 60:1473–75
    [Google Scholar]
  25. 25. 
    Cheng L, You Q, Yin H, Holt MP, Ju C 2010. Involvement of natural killer T cells in halothane-induced liver injury in mice. Biochem. Pharmacol. 80:255–61
    [Google Scholar]
  26. 26. 
    Akai S, Hosomi H, Minami K, Tsuneyama K, Katoh M et al. 2007. Knock down of γ-glutamylcysteine synthetase in rat causes acetaminophen-induced hepatotoxicity. J. Biol. Chem. 282:23996–4003
    [Google Scholar]
  27. 27. 
    Morita M, Akai S, Hosomi H, Tsuneyama K, Nakajima M, Yokoi T 2009. Drug-induced hepatotoxicity test using γ-glutamylcysteine synthetase knockdown rat. Toxicol. Lett. 189:159–65
    [Google Scholar]
  28. 28. 
    Yoshikawa Y, Morita M, Hosomi H, Tsuneyama K, Fukami T et al. 2009. Knockdown of superoxide dismutase-2 enhances acetaminophen-induced hepatotoxicity in rat. Toxicology 264:89–95
    [Google Scholar]
  29. 29. 
    Liu M, Zhao Y, Zhang X 2015. Knockdown of glutamate cysteine ligase catalytic subunit by siRNA causes the gold nanoparticles-induced cytotoxicity in lung cancer cells. PLOS ONE 10:e0118870
    [Google Scholar]
  30. 30. 
    Hosomi H, Akai S, Minami K, Yoshikawa Y, Fukami T et al. 2010. An in vivo drug-induced hepatotoxicity screening system using CYP3A4-expressing and γ-glutamylcysteine synthetase knockdown cells. Toxicol. In Vitro 24:1032–38
    [Google Scholar]
  31. 31. 
    Yoshikawa Y, Hosomi H, Fukami T, Nakajima M, Yokoi T 2009. Establishment of knockdown of superoxide dismutase 2 and expression of CYP3A4 cell system to evaluate drug-induced cytotoxicity. Toxicol. In Vitro 23:1179–87
    [Google Scholar]
  32. 32. 
    Lee YH, Chung MC, Lin Q, Boelsterli UA 2008. Troglitazone-induced hepatic mitochondrial proteome expression dynamics in heterozygous Sod2+/− mice: two-stage oxidative injury. Toxicol. Appl. Pharmacol. 231:43–51
    [Google Scholar]
  33. 33. 
    Lu SC. 2013. Glutathione synthesis. Biochim. Biophys. Acta 1830:3143–53
    [Google Scholar]
  34. 34. 
    Griffith OW. 1982. Mechanism of action, metabolism, and toxicity of buthionine sulfoximine and its higher homologs, potent inhibitors of glutathione synthesis. J. Biol. Chem. 257:13704–12
    [Google Scholar]
  35. 35. 
    Kiyosawa N, Uehara T, Gao W‐H, Omura K, Hirode M et al. 2007. Identification of glutathione depletion-responsive genes using phorone‐treated rat liver. J. Toxicol. Sci. 32:469–86
    [Google Scholar]
  36. 36. 
    Gao W, Mizukawa Y, Nakatsu N, Minowa Y, Yamada H et al. 2010. Mechanism-based biomarker gene sets for glutathione depletion-related hepatotoxicity in rats. Toxicol. Appl. Pharmacol. 247:211–21
    [Google Scholar]
  37. 37. 
    Iida A, Sasaki E, Oda S, Yano A, Tsuneyama K et al. 2015. Carbamazepine-induced liver injury required CYP3A-mediated metabolism and glutathione depletion in rats. Drug Metab. Dispos. 43:958–68
    [Google Scholar]
  38. 38. 
    Allameh A, Vansoun EY, Zarghi A 1997. Role of glutathione conjugation in protection of weanling rat liver against acetaminophen-induced hepatotoxicity. Mech. Ageing Dev. 95:71–79
    [Google Scholar]
  39. 39. 
    Bailey MJ, Dickinson RG. 2003. Acyl glucuronide reactivity in perspective: biological consequences. Chem. Biol. Interact. 145:117–37
    [Google Scholar]
  40. 40. 
    Boelsterli UA. 2002. Xenobiotic acyl glucuronides and acyl CoA thioesters as protein-reactive metabolites with the potential to cause idiosyncratic drug reactions. Curr. Drug Metab. 3:439–50
    [Google Scholar]
  41. 41. 
    Sawamura R, Okudaira N, Watanabe K, Murai T, Kobayashi Y et al. 2010. Predictability of idiosyncratic drug toxicity risk for carboxylic acid containing drugs based on the chemical stability of acyl glucuronide. Drug Metab. Dispos. 38:1857–64
    [Google Scholar]
  42. 42. 
    Jinno N, Ohashi S, Tagashira M, Kohira T, Yamada S 2013. A simple method to evaluate reactivity of acylglucuronides optimized for early stage drug discovery. Biol. Pharm. Bull. 36:1509–13
    [Google Scholar]
  43. 43. 
    Iwamura A, Ito M, Mitsui H, Hasegawa J, Kosaka K et al. 2015. Toxicological evaluation of acyl glucuronides utilizing half-lives, peptide adducts, and immunostimulation assays. Toxicol. In Vitro 30:241–49
    [Google Scholar]
  44. 44. 
    Wang J, Davis M, Li F, Azam F, Scatina J, Talaat R 2004. 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. 17:1206–16
    [Google Scholar]
  45. 45. 
    Miyashita T, Kimura K, Fukami T, Nakajima M, Yokoi T 2014. Evaluation and mechanistic analysis of the cytotoxicity of the acyl glucuronide of nonsteroidal anti-inflammatory drugs. Drug Metab. Dispos. 42:1–8
    [Google Scholar]
  46. 46. 
    Jiao W, Zhao X, Wu G, Zhang X, Wu H, Cui Y 2020. Bioactivation of lumiracoxib in human liver microsomes: formation of GSH- and amino adducts through acyl glucuronide. Drug Test Anal 12:827–35
    [Google Scholar]
  47. 47. 
    Mitsugi R, Sumida K, Fujie Y, Tukey RH, Itoh T, Fujiwara R 2016. Acyl-glucuronide as a possible cause of trovafloxacin-induced liver toxicity: induction of chemokine (C-X-C motif) ligand 2 by trovafloxacin acyl-glucuronide. Biol. Pharm. Bull. 39:1604–10
    [Google Scholar]
  48. 48. 
    Oda S, Shirai Y, Akai S, Nakajima A, Tsuneyama K, Yokoi T 2016. Toxicological role of an acyl glucuronide metabolite in diclofenac-induced acute liver injury in mice. J. Appl. Toxicol. 37:545–53
    [Google Scholar]
  49. 49. 
    Iwamura A, Nakajima M, Oda S, Yokoi T 2017. Toxicological potential of acyl glucuronides and its assessment. Drug Metabol. Pharmacokinet. 32:2–11
    [Google Scholar]
  50. 50. 
    Buchweitz JP, Ganey PE, Bursian SJ, Roth RA 2002. Underlying endotoxemia augments toxic responses to chlorpromazine: Is there a relationship to drug idiosyncrasy. ? J. Pharmacol. Exp. Ther. 300:460–67
    [Google Scholar]
  51. 51. 
    Deng X, Stachlewitz RF, Liguori MJ, Blomme EAG, Waring JF et al. 2006. Modest inflammation enhances diclofenac hepatotoxicity in rats: role of neutrophils and bacterial translocation. J. Pharmacol. Exp. Ther. 300:1191–99
    [Google Scholar]
  52. 52. 
    Tukov FF, Luyendyk JP, Ganey PE, Roth RA 2007. The role of tumor necrosis factor alpha in lipopolysaccharide/ranitidine-induced inflammatory liver injury. Toxicol. Sci. 100:267–80
    [Google Scholar]
  53. 53. 
    Zou W, Devi SS, Sparkenbaugh E, Younis HS, Roth RA, Ganey PE 2009. Hepatotoxic interaction of sulindac with lipopolysaccharide: role of the hemostatic system. Toxicol. Sci. 108:184–93
    [Google Scholar]
  54. 54. 
    Shaw PJ, Ganey PE, Roth RA 2010. Idiosyncratic drug-induced liver injury and the role of inflammatory stress with an emphasis on an animal model of trovafloxacin hepatotoxicity. Toxicol. Sci. 118:7–18
    [Google Scholar]
  55. 55. 
    Kobayashi M, Higuchi S, Mizuno K, Tsuneyama K, Fukami T et al. 2010. Interleukin-17 is involved in α-naphthylisothiocyanate-induced liver injury in mice. Toxicology 275:50–57
    [Google Scholar]
  56. 56. 
    Yano A, Higuchi S, Tsuneyama K, Fukami T, Nakajima M, Yokoi T 2012. Involvement of immune-related factors in diclofenac-induced acute liver in mice. Toxicology 293:107–14
    [Google Scholar]
  57. 57. 
    Takai S, Higuchi S, Yano A, Tsuneyama K, Fukami T et al. 2015. Involvement of immune- and inflammatory-related factors in flucloxacillin-induced liver injury in mice. J. Appl. Toxicol. 35:142–51
    [Google Scholar]
  58. 58. 
    Higuchi S, Kobayashi M, Yoshikawa Y, Tsuneyama K, Fukami T et al. 2011. IL-4 mediated dicloxacillin-induced liver injury in mice. Toxicol. Lett. 200:139–45
    [Google Scholar]
  59. 59. 
    Higuchi S, Kobayashi M, Yano A, Tsuneyama K, Fukami T et al. 2012. Involvement of Th2 cytokines in the mouse model of flutamide-induced acute liver injury. J. Appl. Toxicol. 32:815–22
    [Google Scholar]
  60. 60. 
    Kobayashi M, Higuchi S, Ide M, Nishikawa S, Fukami T et al. 2012. Th2 cytokines-mediated methimazole-induced liver injury in mice. J. Appl. Toxicol. 32:823–33
    [Google Scholar]
  61. 61. 
    Novartis Pharm. Co. 2011. Interview Form (Product Information Booklet) of Tegretol® Product monograph . , 9th. ed. Novartis Pharma Co Tokyo:
  62. 62. 
    Takao K, Miyakawa T. 2015. Genomic responses in mouse models greatly mimic human inflammatory diseases. PNAS 112:1167–72
    [Google Scholar]
  63. 63. 
    Sasaki E, Yokoi T. 2018. Role of cytochrome P450-mediated metabolism and involvement of reactive metabolite formations on antiepileptic drug-induced liver injuries. J. Toxicol. Sci. 43:75–87
    [Google Scholar]
  64. 64. 
    Li J, Zhu X, Liu F, Cai P, Sanders C, Lee WM et al. 2010. Cytokine and autoantibody patterns in acute liver failure. J. Immunotoxicol. 7:157–64
    [Google Scholar]
  65. 65. 
    Bourdi M, Chen W, Peter RM, Martin JL, Buters JT et al. 1996. Human cytochrome P450 2E1 is a major autoantigen associated with halothane hepatitis. Chem. Res. Toxicol. 9:1159–66
    [Google Scholar]
  66. 66. 
    Maniratanachote R, Shibata A, Kaneko S, Wakasugi T, Sawazaki T et al. 2005. Identification of autoantibody to aldolase B in sera from patients with troglitazone-induced liver disfunction. Toxicology 216:15–23
    [Google Scholar]
  67. 67. 
    Debock I, Flamand V. 2014. Unbalanced neonatal CD4+ T-cell immunity. Front. Immunol. 5:393
    [Google Scholar]
  68. 68. 
    Leung S, Liu X, Fang L, Chen X, Guo T, Zhang J 2010. The cytokine milieu in the interplay of pathogenic Th1/Th17 cells and regulatory T cells in autoimmune disease. Cell. Mol. Immunol. 7:182–89
    [Google Scholar]
  69. 69. 
    Zhou L, Ivanov II, Spolski R, Min R, Shenderov K et al. 2007. IL-6 programs Th-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. 8:967–74
    [Google Scholar]
  70. 70. 
    Daly AK, Donaldson PT, Bhatnagar P, Shen Y, Pe'er I et al. 2009. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat. Genet. 41:816–19
    [Google Scholar]
  71. 71. 
    Hirata K, Takagi H, Yamamoto M, Matsumoto T, Nishiya T et al. 2008. Ticlopidine-induced hepatotoxicity is associated with specific human leukocyte antigen genomic subtypes in Japanese patients: a preliminary case-control study. Pharmacogenomics J 8:29–33
    [Google Scholar]
  72. 72. 
    Hautekeete ML, Horsmans Y, Van Waeyenberge C, Demanet C, Henrion J et al. 1999. HLA association of amoxicillin-clavulanate-induced hepatitis. Gastroenterology 117:1181–86
    [Google Scholar]
  73. 73. 
    Kindmark A, Jawaid A, Harbron CG, Barratt BJ, Bengtsson OF et al. 2008. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J 8:186–95
    [Google Scholar]
  74. 74. 
    Singer JB, Lewitzky S, Leroy E, Yang F, Zhao X et al. 2010. A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat. Genet. 42:711–14
    [Google Scholar]
  75. 75. 
    Walgren JL, Mitchell MD, Thompson DC 2005. Role of metabolism in drug-induced idiosyncratic hepatotoxicity. Crit. Rev. Toxicol. 35:325–61
    [Google Scholar]
  76. 76. 
    Usui T, Mise M, Hashizume T, Yabuki M, Komuro S 2009. Evaluation of the potential for drug-induced liver injury based on in vitro covalent binding to human liver proteins. Drug Metab. Dispos. 37:2383–92
    [Google Scholar]
  77. 77. 
    Evans DC, Watt AP, Nicoll-Griffith DA, Baillie TA 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]
  78. 78. 
    Nakayama S, Atsumi R, Takakusa H, Kobayashi Y, Kurihara A 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]
  79. 79. 
    Persson M, Løye AF, Jacquet M, Mow NS, Thougaard AV et al. 2014. High-content analysis/screening for predictive toxicology: application to hepatotoxicity and genotoxicity. Basic Clin. Pharmacol. Toxicol. 115:18–23
    [Google Scholar]
  80. 80. 
    Martin HL, Adams M, Higgins J, Bond J, Morrison EE et al. 2014. High-content, high-throughput screening for the identification of cytotoxic compounds based on cell morphology and cell proliferation markers. PLOS ONE 9:e88338
    [Google Scholar]
  81. 81. 
    Aninat C, Piton A, Glaise D, Le Charpentier T, Langouët S et al. 2006. Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug Metab. Dispos. 34:75–83
    [Google Scholar]
  82. 82. 
    Xu J, Oda S, Yokoi T 2018. Cell-based assay using glutathione-depleted HepaRG human liver cells for predicting drug-induced liver injury. Toxicol. In Vitro 48:286–301
    [Google Scholar]
  83. 83. 
    Nishimura M, Ejiri Y, Kishomoto S, Suzuki S, Satoh T et al. 2011. Expression levels of drug-metabolizing enzyme, transporter, and nuclear receptor mRNAs in a novel three-dimensional culture system for human hepatocytes using micro-space plates. Drug Metab. Pharmacokinet. 26:137–44
    [Google Scholar]
  84. 84. 
    Tomida T, Okamura H, Yokoi T, Konno Y 2017. A modified multiparametric assay using HepaRG cells for predicting the degree of drug-induced liver injury risk. J. Appl. Toxicol. 37:382–90
    [Google Scholar]
  85. 85. 
    Liu ZX, Govindarajan S, Kaplowitz N 2004. Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity. Gastroenterology 127:1760–74
    [Google Scholar]
  86. 86. 
    Jaeschke H. 2005. Role of inflammation in the mechanism of acetaminophen-induced hepatotoxicity. Expert Opin. Drug Metab. Toxicol. 1:389–97
    [Google Scholar]
  87. 87. 
    Granitzny A, Knebel J, Müller M, Braun A, Steinberg P et al. 2017. Evaluation of a human in vitro hepatocyte-NPC co-culture model for the prediction of idiosyncratic drug-induced liver injury: a pilot study. Toxicol. Rep. 4:89–103
    [Google Scholar]
  88. 88. 
    Kato R, Uetrecht J. 2017. Supernatant from hepatocyte cultures with drugs that cause idiosyncratic liver injury activates macrophage inflammasomes. Chem. Res. Toxicol. 30:1327–32
    [Google Scholar]
  89. 89. 
    Mak A, Kato R, Weston K, Hayes A, Uetrecht J 2018. An impaired immune tolerance animal model distinguishes the potential of troglitazone/pioglitazone and tolcapone/entacapone to cause IDILI. Toxicol. Sci. 161:412–20
    [Google Scholar]
  90. 90. 
    Yano A, Oda S, Fukami T, Nakajima M, Yokoi T 2014. Development of a cell-based assay system considering drug metabolism and immune- and inflammatory-related factors for the risk assessment of drug-induced liver injury. Toxicol. Lett. 228:13–24
    [Google Scholar]
  91. 91. 
    McGill MR, Yan H-M, Ramachandran A, Murray GJ, Rollins DE, Jaeschke H 2011. HepaRG cells: a human model to study mechanisms of acetaminophen hepatotoxicity. Hepatology 53:974–82
    [Google Scholar]
  92. 92. 
    Ponsoda X, Bort R, Jover R, Gómez-Lechón MJ, Castell JV 1999. Increased toxicity of cocaine on human hepatocytes induced by ethanol: role of GSH. Biochem. Pharmacol. 58:1579–85
    [Google Scholar]
  93. 93. 
    Stepan AF, Walker DP, Bauman J, Price DA, Baillie TA 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]
  94. 94. 
    Yuan L, Kaplowitz N. 2009. Glutathione in liver diseases and hepatotoxicity. Mol. Asp. Med. 30:29–41
    [Google Scholar]
  95. 95. 
    O'Brien PJ, Irwin W, Diaz D, Howard-Cofield E, Krejsa CM et al. 2006. High concordance of drug-induced human hepatotoxicity with in vitro cytotoxicity measured in a novel cell-based model using high content screening. Arch. Toxicol. 80:580–604
    [Google Scholar]
  96. 96. 
    Liguori MJ, Ditewig AC, Maddox JF, Luyendyk JP, Lehman-McKeeman LD et al. 2010. Comparison of TNFα to lipopolysaccharide as an inflammagen to characterize the idiosyncratic hepatotoxicity potential of drugs: trovafloxacin as an example. Int. J. Mol. Sci. 11:4697–714
    [Google Scholar]
  97. 97. 
    Song B, Aoki S, Liu C, Ito K 2019. A Toll-like receptor 9 agonist sensitizes mice to mitochondrial dysfunction-induced hepatic apoptosis via the Fas/FasL pathway. Arch. Toxicol. 93:1573–84
    [Google Scholar]
  98. 98. 
    Edling Y, Sivertsson LK, Butura A, Ingelman-Sundberg M, Ek M 2009. Increased sensitivity for troglitazone-induced cytotoxicity using a human in vitro co-culture model. Toxicol. In Vitro 23:1387–95
    [Google Scholar]
  99. 99. 
    Kim DE, Jang M-J, Kim YR, Lee J-Y, Cho EB et al. 2017. Prediction of drug-induced immune-mediated hepatotoxicity using hepatocyte-like cells derived from human embryonic stem cells. Toxicology 387:1–9
    [Google Scholar]
  100. 100. 
    Oda S, Uchida Y, Aleo MD, Koza-Taylor PH, Matsui Y et al. 2020. An in vitro coculture system of human peripheral blood mononuclear cells with hepatocellular carcinoma-derived cells for predicting drug-induced liver injury. Arch. Toxicol. In press
    [Google Scholar]
  101. 101. 
    Ter Horst R, Jaeger M, Smeekens SP, Oosting M, Swertz MA et al. 2016. Host and environmental factors influencing individual human cytokine responses. Cell 167:1111–24
    [Google Scholar]
  102. 102. 
    Wang K, Zhang S, Marzolf B, Troisch P, Brightman A et al. 2009. Circulating microRNAs, potential biomarkers for drug-induced liver injury. PNAS 106:4402–7
    [Google Scholar]
  103. 103. 
    He L, Hannon GL. 2004. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 5:522–31
    [Google Scholar]
  104. 104. 
    Esteller M. 2011. Non-coding RNAs in human disease. Nat. Rev. Genet. 12:861–74
    [Google Scholar]
  105. 105. 
    Kagawa T, Shirai Y, Oda S, Yokoi T 2018. Identification of specific microRNA biomarkers in early stages of hepatocellular injury, cholestasis, and steatosis in rats. Toxicol. Sci. 166:228–39
    [Google Scholar]
  106. 106. 
    Yamaura Y, Nakajima M, Takagi S, Fukami T, Tsuneyama K, Yokoi T 2012. Plasma microRNA profiles in rat models of hepatocellular injury, cholestasis, and steatosis. PLOS ONE 7:e30250
    [Google Scholar]
  107. 107. 
    Carissimi C, Fulci V, Macino G 2009. MicroRNAs: novel regulators of immunity. Autoimmune Rev 8:520–24
    [Google Scholar]
  108. 108. 
    Endo S, Yano A, Fukami T, Nakajima M, Yokoi T 2014. Involvement of miRNAs in the early phase of halothane-induced liver injury. Toxicology 319:75–84
    [Google Scholar]
  109. 109. 
    Uematsu Y, Akai S, Tochitani T, Tateishi Y, Oda S, Yokoi T 2016. Micro RNA-mediated Th2 bias in the pathogenesis of methimazole-induced acute liver injury in mice. Toxicol. Appl. Pharmacol. 307:1–9
    [Google Scholar]
  110. 110. 
    Sanjay S, Girish C. 2017. Role of miRNA and its potential as a novel diagnostic biomarker in drug-induced liver injury. Eur. J. Clin. Pharmacol. 73:399–407
    [Google Scholar]
  111. 111. 
    Li Y, Ren Q, Zhu L, Li Y, Li J et al. 2018. Involvement of methylation of microRNA-122, -125b and 106b in regulation of cyclin G1, CAT-1 and STAT3 target genes in isoniazid-induced liver injury. BMC Pharmacol. Toxicol. 19:11
    [Google Scholar]
  112. 112. 
    Physicians’ Desk Reference. 2000. Rezulin. Physicians’ Desk Reference PDR Staff 2310–14 Rutherford, NJ: Med. Econ. Co. , 53rd. ed.
    [Google Scholar]
  113. 113. 
    Kassahun K, Person PG, Tang W, McIntosh I, Leung K et al. 2001. 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. 14:62–70
    [Google Scholar]
  114. 114. 
    Tettey JN, Maggs JL, Rapeport WG, Pirmohamed M, Park BK 2001. Enzyme-induction dependent bioactivation of troglitazone and troglitazone quinone in vivo. Chem. Res. Toxicol. 14:965–74
    [Google Scholar]
  115. 115. 
    Yamamoto Y, Yamazaki Y, Ikeda T, Watanabe T, Iwabuchi H et al. 2002. Formation of a novel epoxide form of quinone metabolite of troglitazone with cytotoxicity to HepG2 cells. Drug Metab. Dispos. 30:155–60
    [Google Scholar]
  116. 116. 
    Vignati L, Turlizzi E, Monaci S, Grossi P, de Kanter R, Monshouwer M 2005. An in vitro approach to detect metabolite toxicity due to CYP3A4-dependent bioactivation of xenobiotics. Toxicology 216:154–67
    [Google Scholar]
  117. 117. 
    Watanabe T, Ohashi Y, Yasuda M, Takaoka M, Furukawa T et al. 1999. Was it not possible to predict liver dysfunction caused by troglitazone during the nonclinical safety studies? Reevaluation of safety. Iyakuhin Kenkyu 30:537–46
    [Google Scholar]
  118. 118. 
    Fujimoto K, Kumagai K, Ito K, Arakawa S, Ando Y et al. 2009. Sensitivity of liver injury in heterozygous Sod2 knockout mice treated with troglitazone or acetaminophen. Toxicol. Pathol. 37:193–200
    [Google Scholar]
  119. 119. 
    Tateno C, Yoshizane Y, Saito N, Kataoka M, Utoh R et al. 2004. Near completely humanized liver in mice shows human-type metabolic responses to drugs. Am. J. Pathol. 165:901–12
    [Google Scholar]
  120. 120. 
    Kakuni M, Morita M, Matsuo K, Katoh Y, Nakajima M et al. 2012. Chimeric mice with a humanized liver as an animal model of troglitazone-induced liver injury. Toxicol. Lett. 214:9–18
    [Google Scholar]
  121. 121. 
    Jia R, Oda S, Tsuneyama K, Urano Y, Yokoi T 2019. Establishment of a mouse model of troglitazone-induced liver injury and analysis of its hepatotoxic mechanism. J. Appl. Toxicol. 39:1541–56
    [Google Scholar]
  122. 122. 
    Sasaki E, Iida A, Oda S, Tsuneyama K, Fukami T et al. 2016. Pathogenetic analyses of carbamazepine-induced liver injury in F344 rats focused on morphology and immune- and inflammation-related factors. Exp. Toxicol. Pathol. 68:27–38
    [Google Scholar]
  123. 123. 
    Akai S, Uematsu Y, Tsuneyama K, Oda S, Yokoi T 2016. Kupffer cell-mediated exacerbation of methimazole-induced acute liver injury in rats. J. Appl. Toxicol. 36:702–15
    [Google Scholar]
  124. 124. 
    Mazurek SR, Bovo E, Zima AV 2014. Regulation of sarcoplasmic reticulum Ca2+ release by cytosolic glutathione in rabbit ventricular myocytes. Free Radical Biol. Med. 68:159–67
    [Google Scholar]
  125. 125. 
    Giorgi C, De Stefani D, Bononi A, Rizzuto R, Pinton P 2009. Structural and functional link between the mitochondrial network and the endoplasmic reticulum. Int. J. Biochem. Cell Biol. 41:1817–27
    [Google Scholar]
  126. 126. 
    Takai S, Oda S, Tsuneyama K, Fukami T, Nakajima M, Yokoi T 2016. Establishment of a mouse model for amiodarone-induced liver injury and analyses of its hepatotoxic mechanism. J. Appl. Toxicol. 36:35–37
    [Google Scholar]
  127. 127. 
    Shirai Y, Oda S, Makino S, Tsuneyama K, Yokoi T 2017. Establishment of a mouse model of enalapril-induced liver injury and investigation of a pathogenesis. Lab. Investig. 97:833–42
    [Google Scholar]
  128. 128. 
    Urano Y, Oda S, Tsuneyama K, Yokoi T 2018. Comparative hepatic transcript analyses revealed possible pathogenic mechanisms of fasiglifam (TAK-875)-induced acute liver injury in mice. Chem. Biol. Interact. 296:185–97
    [Google Scholar]
  129. 129. 
    Akai S, Oda S, Yokoi T 2018. Strain and interindividual differences in lamotrigine-induced liver injury in mice. J. Appl. Toxicol. 39:451–60
    [Google Scholar]
  130. 130. 
    Matsuo K, Sasaki E, Higuchi S, Takai S, Tsuneyama K et al. 2014. Involvement of oxidative stress and immune- and inflammation-related factors in azathioprine-induced liver injury. Toxicol. Lett. 224:215–24
    [Google Scholar]
  131. 131. 
    Hossain MB, Hosokawa H, Hasegawa A, Watarai H, Taniguchi M et al. 2008. Lymphoid enhancer factor interacts with GATA-3 and controls its function in T helper type 2 cells. Immunology 125:377–86
    [Google Scholar]
  132. 132. 
    Kuwahara M, Yamashita M, Shinoda K, Tofukuji S, Onodera A et al. 2012. The transcription factor Sox4 is a downstream target of signaling by the cytokine TGF-β and suppresses TH2 differentiation. Nat. Immunol. 13:778–86
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-030220-015007
Loading
/content/journals/10.1146/annurev-pharmtox-030220-015007
Loading

Data & Media loading...

Supplemental Material

Supplementary Data

  • 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