Immune mechanisms of idiosyncratic drug-induced liver injury

Idiosyncratic drug reactions (IDRs) continue to be an important issue. Specifically, idiosyncratic drug-induced liver injury (IDILI) is the most likely IDR to lead to drug withdrawal, and it accounts for a significant portion of all cases of acute liver failure. In addition, IDRs are unpredictable and their mechanisms are not well understood. There is increasing clinical evidence that most IDILI is immune mediated. Several immune mediated mechanistic hypotheses exist such as the hapten and danger hypothesis; however, they do not completely explain the idiosyn cratic nature of these reactions. Extensive mechanistic studies are needed to better understand these reactions; however, it is impossible to do controlled experiments in humans, and previous animal models did not properly model IDILI. If IDILI is immune mediated and the major factor preventing liver injury in patients is immune tolerance, then a plausible method to develop an animal model of IDILI would be to impair immune tolerance. This hypothesis has shown promise in developing valid animal models of IDILI as demonstrated by a halothane induced liver injury mouse model developed by depleting myeloid derived suppressor cells (MDSCs), as well as an amodiaquine-, isoniazid- and nevirapine-induced liver injury mouse model developed by impairing immune tolerance by blocking PD-1 and CTLA-4, two immune checkpoint inhibitors. Further characterization and valida tion of these models is required; however, it is likely that they will make it possible to perform mechanistic studies that have been impossible in the past. Relevance for patients: Idiosyncratic drug-induced liver injury can be serious leading to liver transplantation or death. Their idiosyncratic nature makes mechanistic studies very difficult. However, with the development of the first animal model that is similar to the liver injury that occurs in humans, it will be possible to study the mecha nisms involved. With a better mechanistic understanding it should be possible to test drug candidates and pr
[1] Uetrecht J, Naisbitt DJ. Idiosyncratic adverse drug reactions: current concepts. Pharmacol Rev 2013; 65: 779-808.
[2] Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, Farrar K, Park BK, Breckenridge AM. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ 2004; 329, 15-19.
[3] Hunziker T, Bruppacher R, Kuenzi UP, Maibach R, Braunschweig S, Halter F, Hoigne RV. Classification of ADRs: aproposal for harmonization and differentiation based on the experience of the Comprehensive Hospital Drug Monitoring Bern/St. Gallen, 1974-1993. Pharmacoepidemiol Drug Saf 2002; 11: 159-163.
[4] Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. JAMA 2002; 287: 2215-2220.
[5] Uetrecht, J. Idiosyncratic drug reactions: current understanding. Annu Rev Pharmacol Toxicol 2007; 47: 513-539.
[6] Chalasani NP, Hayashi PH, Bonkovsky HL, Navarro VJ, Lee WM, Fontana RJ, Practice Parameters Committee of the American College of, G. ACG Clinical Guideline: the diagnosis and management of idiosyncratic drug-induced liver injury. Am J Gastroenterol 2014; 109: 950-966; quiz 967.
[7] Onakpoya IJ, Heneghan CJ, Aronson JK. Post-marketing withdrawal of 462 medicinal products because of adverse drug reactions: a systematic review of the world literature. BMC Med 2016; 14: 10.
[8] Ostapowicz G, Fontana RJ, Schiodt FV, Larson A, Davern TJ, Han SH, McCashland TM, Shakil AO, Hay JE, Hynan L, Crippin JS, Blei AT, Samuel G, Reisch J, Lee WM, Group U.S.A.L.F.S. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 2002; 137, 947-954.
[9] Singh D, Cho WC, Upadhyay G. Drug-induced liver toxicity and prevention by herbal antioxidants: an overview. Front Physiol 2015; 6: 363.
[10] Medzhitov R, Janeway CA Jr. Innate immunity: the virtues of a nonclonal system of recognition. Cell 1997; 91: 295-298.
[11] Foureau DM, Walling TL, Maddukuri V, Anderson W, Culbreath K, Kleiner DE, Ahrens WA, Jacobs C, Watkins PB, Fontana RJ, Chalasani N, Talwalkar J, Lee WM, Stolz A, Serrano J, Bonkovsky HL. Comparative analysis of portal hepatic infiltrating leucocytes in acute drug-induced liver injury, idiopathic autoimmune and viral hepatitis. Clin Exp Immunol 2015; 180: 40-51.
[12] Greaves RR, Agarwal A, Patch D, Davies SE, Sherman D, Reynolds N, Rolles K, Davidson BR, Burroughs AK. Inadvertent diclofenac rechallenge from generic and non-generic prescribing, leading to liver transplantation for fulminant liver failure. Eur J Gastroenterol Hepatol 2001; 13: 71-73.
[13] Papay JI, Clines D, Rafi R, Yuen N, Britt SD, Walsh JS, Hunt CM. Drug-induced liver injury following positive drug rechallenge. Regul Toxicol Pharmacol 2009; 54: 84-90.
[14] Warkentin TE, Kelton JG. Temporal aspects of heparin-induced thrombocytopenia. N Engl J Med 2001; 344: 1286-1292.
[15] Lee WM, Larrey D, Olsson R, Lewis JH, Keisu M, Auclert L, Sheth S. Hepatic findings in long-term clinical trials of ximelagatran. Drug Saf 2005; 28: 351-370.
[16] Tahaoğlu K, Ataç G, Sevim T, Tärün T, Yazicioğlu O, Horzum G, Gemci I, Ongel A, Kapakli N, Aksoy E. The management of anti-tuberculosis drug-induced hepatotoxicity. Int J Tuberc Lung Dis 2001; 5: 65-69.
[17] Warrington RJ, Tse KS, Gorski BA, Schwenk R, Sehon AH.Evaluation of isoniazid-associated hepatitis by immunological tests. Clin Exp Immunol 1978; 32: 97-104.
[18] Nyfeler B, Pichler WJ. The lymphocyte transformation test for the diagnosis of drug allergy: sensitivity and specificity. Clin Exp Allergy 1997; 27: 175-181.
[19] Warrington RJ, McPhilips-Feener S, Rutherford W. The predictive value of the lymphocyte transformation test in isoniazid-associated hepatitis. Clin Exp Allergy 1982; 12: 217−222.
[20] Liu ZX, Kaplowitz N. Immune-mediated drug- induced liver disease. Clin Liver Dis 2002; 6: 755-774.
[21] Metushi IG, Sanders C, Acute Liver Study G, Lee WM, Uetrecht J. Detection of anti-isoniazid and anti-cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Hepatology 2014; 59: 1084-1093.
[22] Mallal S, Phillips E, Carosi G, Molina JM, Workman C, Tomazic J, Jagel-Guedes E, Rugina S, Kozyrev O, Cid JF, Hay P, Nolan D, Hughes S, Hughes A, Ryan S, Fitch N, Thorborn D, Benbow A, Team PS. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008; 358: 568-579.
[23] Mallal S, Nolan D, Witt C, Masel G, Martin AM, Moore C, Sayer D, Castley A, Mamotte C, Maxwell D, James I, Christiansen FT. Association between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to HIV-1 reverse-transcriptase inhibitor abacavir. Lancet 2002; 359: 727-732.
[24] Daly AK, Donaldson PT, Bhatnagar P, Shen Y, Pe'er I, Floratos A, Daly MJ, Goldstein DB, John S, Nelson MR, Graham J, Park BK, Dillon JF, Bernal W, Cordell HJ, Pirmohamed M, Aithal GP, Day CP; DILIGEN Study; International SAE Consortium. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet 2009; 41: 816-819.
[25] Kindmark A, Jawaid A, Harbron CG, Barratt BJ, Bengtsson OF, Andersson TB, Carlsson S, Cederbrant KE, Gibson NJ, Armstrong M, Lagerstrom-Fermer ME, Dellsen A, Brown EM, Thornton M, Dukes C, Jenkins SC, Firth MA, Harrod GO, Pinel TH, Billing-Clason SM, Cardon LR, March RE. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J 2008; 8: 186-195.
[26] Donaldson PT, Daly AK, Henderson J, Graham J, Pirmohamed M, Bernal W, Day CP, Aithal GP. Human leucocyte antigen class II genotype in susceptibility and resistance to co-amoxiclav-induced liver injury. J Hepatol 2010; 53: 1049-1053.
[27] Sharma SK, Balamurugan A, Saha PK, Pandey RM, Mehra NK. Evaluation of clinical and immunogenetic risk factors for the development of hepatotoxicity during antituberculosis treatment. Am J Respir Crit Care Med 2002; 166: 916-919.
[28] Metushi IG, Uetrecht J, Phillips E. Mechanism of isoniazid-induced hepatotoxicity: then and now. Br J Clin Pharmacol 2016; 81: 1030-1036.
[29] Landsteiner K, Jacobs J. Studies on the Sensitization of Animals with Simple Chemical Compounds. J Exp Med 1935; 61: 643-656.
[30] Uetrecht J. Prediction of a new drug's potential to cause idiosyncratic reactions. Curr Opin Drug Discov Devel 2001; 4: 55-59.
[31] Metushi IG, Nakagawa T, Uetrecht J. Direct oxidation and covalent binding of isoniazid to rodent liver and human hepatic microsomes: humans are more like mice than rats. Chem Res Toxicol 2012; 25: 2567-2576.
[32] Metushi IG, Cai P, Dervovic D, Liu F, Lobach A, Nakagawa T, Uetrecht J. Development of a novel mouse model of amodiaquine-induced liver injury with a delayed onset. J Immunotoxicol 2015; 12: 247-260.
[33] Sharma AM, Li Y, Novalen M, Hayes MA, Uetrecht J. Bioactivation of nevirapine to a reactive quinone methide: implications for liver injury. Chem Res Toxicol 2012; 25: 1708-1719.
[34] Matzinger P. Tolerance, danger, and the extended family. Ann Rev Immunol 1994; 12: 991-1045.
[35] Gallucci S, Matzinger P. Danger signals: SOS to the immune system. Curr Opin Immunol 2001; 13: 114-119.
[36] Uetrecht J, Li J, Kourteva G, Hilton H, Cai P, Workman HC, Haggerty H, Moore D. Changes in Hepatic Gene Expression Induced by Amodiaquine in Rats. In: 2011 Annual Meeting Abstract, Society of Toxicology. Abstract no. 1503.
[37] Zimmerman HJ. Hepatotoxicity: the adverse effects of drugs and other chemicals on the liver. Lippincott Williams & Wilkins. 1999.
[38] Luyendyk JP, Maddox JF, Cosma GN, Ganey PE, Cockerell GL, Roth RA. Ranitidine treatment during a modest inflammatory response precipitates idiosyncrasy-like liver injury in rats. J Pharmacol Exp Ther 2003; 307: 9-16.
[39] You Q, Cheng L, Ju C. Generation of T cell responses targeting the reactive metabolite of halothane in mice. Toxicol Lett 2010; 194: 79-85.
[40] Mak A, Uetrecht J. The role of CD8 T cells in amodiaquineinduced liver injury in PD-1-/- mice cotreated with antiCTLA-4. Chem Res Toxicol 2015; 28: 1567-1573.
[41] Depta JP, Pichler WJ. Cross-reactivity with drugs at the T cell level. Curr Opin Allergy Clin Immunol 2003; 3: 261-267.
[42] Welsh RM, Selin LK. No one is naive: the significance of heterologous T-cell immunity. Nat Rev Immunol 2002; 2: 417-426.
[43] Meylan E, Tschopp J, Karin M. Intracellular pattern recognition receptors in the host response. Nature 2006; 442: 39-44.
[44] Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med 2015; 21: 677-687.
[45] Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010; 140: 805-820.
[46] Broz P, von Moltke J, Jones JW, Vance RE, Monack DM. Differential requirement for Caspase-1 autoproteolysis in pathogen-induced cell death and cytokine processing. Cell Host Microbe 2010; 8: 471-483.
[47] Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015; 526: 660-665.
[48] Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, Hoffman HM, Feldstein AE. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014; 59: 898-910.
[49] Weston JK, Uetrecht J. Activation of inflammasomes by agents causing idiosyncratic skin reactions: a possible biomarker. Chem Res Toxicol 2014; 27: 949-951.
[50] Meyer UA, Zanger UM. Molecular mechanisms of genetic polymorphisms of drug metabolism. Ann Rev Pharmacol Toxicol 1997; 37: 269-296.
[51] Chakraborty M, Fullerton AM, Semple K, Chea LS, Proctor WR, Bourdi M, Kleiner DE, Zeng X, Ryan PM, Dagur PK, Berkson JD, Reilly TP, Pohl LR. Drug-induced allergic hepatitis develops in mice when myeloid-derived suppressor cells are depleted prior to halothane treatment. Hepatology 2015; 62: 546-557.
[52] Brody GL, Sweet RB. Halothane anesthesia as a possible cause of massive hepatic necrosis. Anesthesiology 1963; 24: 29-37.
[53] Vergani D, Mieli-Vergani G, Alberti A, Neuberger J, Eddleston AL, Davis M, Williams R. Antibodies to the surface of halothane-altered rabbit hepatocytes in patients with severe halothane-associated hepatitis. N Engl J Med 1980; 303: 66-71.
[54] Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9: 162-174.
[55] AlKadi HO. Antimalarial drug toxicity: a review. Chemotherapy 2007; 53: 385-391.
[56] Ng W, Lobach AR, Zhu X, Chen X, Liu F, Metushi IG, Sharma A, Li J, Cai P, Ip J, Novalen M, Popovic M, Zhang X, Tanino T, Nakagawa T, Li Y, Uetrecht J. Animal models of idiosyncratic drug reactions. Adv Pharmacol 2012; 63: 81-135.
[57] Li XQ, Bjorkman A, Andersson TB, Ridderstrom M, Masimirembwa CM. Amodiaquine clearance and its metabolism to N-desethylamodiaquine is mediated by CYP2C8: a new high affinity and turnover enzyme-specific probe substrate. J Pharmacol Exp Ther 2002; 300: 399-407.
[58] Maggs JL, Tingle MD, Kitteringham NR, Park BK. Drugprotein conjugates--XIV. Mechanisms of formation of proteinarylating intermediates from amodiaquine, a myelotoxin and hepatotoxin in man. Biochem Pharmacol 1988; 37: 303-311.
[59] Neftel KA, Woodtly W, Schmid M, Frick PG, Fehr J. Amodiaquine induced agranulocytosis and liver damage. BMJ 1986; 292: 721-723.
[60] Parry RV, Chemnitz JM, Frauwirth KA, Lanfranco AR, Braunstein I, Kobayashi SV, Linsley PS, Thompson CB, Riley JL. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol Cell Biol 2005; 25: 9543-9553.
[61] Das R, Verma R, Sznol M, Boddupalli CS, Gettinger SN, Kluger H, Callahan M, Wolchok JD, Halaban R, Dhodapkar MV, Dhodapkar KM. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J Immunol 2015; 194: 950-959.
[62] Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and itsligands in tolerance and immunity. Ann Rev Immunol 2008; 26: 677-704.
[63] Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252-264.
[64] Walker LS, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol 2011; 11: 852-863.
[65] Egen JG, Kuhns MS, Allison JP. CTLA-4: new insights into its biological function and use in tumor immunotherapy. Nat Immunol 2002; 3: 611-618.
[66] van der Merwe PA, Bodian DL, Daenke S, Linsley P, Davis SJ. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med 1997; 185: 393-403.
[67] Montler R, Bell RB, Thalhofer C, Leidner R, Feng Z, Fox BA, Cheng AC, Bui TG, Tucker C, Hoen H, Weinberg A. OX40, PD-1 and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and neck cancer. Clin Transl Immunology 2016; 5: e70.
[68] Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, Drake CG. Human prostate-infiltrating CD8+ T lymphocytes are oligoclonal and PD-1+. Prostate 2009; 69: 1694-1703.
[69] Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009; 114: 1537- 1544.
[70] Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, Lennon VA, Celis E, Chen L. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002; 8: 793-800.
[71] Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res 2004; 10: 5094-5100.
[72] Cho H, Kang H, Kim CW, Kim HY, Jang JW, Yoon S K, Lee CD. Phenotypic characteristics of PD-1 and CTLA-4 expression in symptomatic acute hepatitis A. Gut Liver 2016; 10: 288-294.
[73] Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ, Walker BD. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006; 443: 350-354.
[74] Nakamoto N, Cho H, Shaked A, Olthoff K, Valiga ME, Kaminski M, Gostick E, Price DA, Freeman GJ, Wherry EJ, Chang KM. Synergistic reversal of intrahepatic HCV-specific CD8 T cell exhaustion by combined PD-1/CTLA-4 blockade. PLoS Pathogens 2009; 5: e1000313.
[75] Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, Flavell RA, Mehal WZ. Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9and the Nalp3 inflammasome. J Clin Invest 2009; 119: 305- 314.
[76] Luis PB, Ruiter J, L IJ, de Almeida IT, Duran M, Wanders RJ, Silva MF. Valproyl-CoA inhibits the activity of ATP- and GTP-dependent succinate:CoA ligases. J Inherit Metab Dis 2014; 37: 353-357.
[77] Martin-Murphy BV, Holt MP, Ju C. The role of damage associated molecular pattern molecules in acetaminophen-inducedliver injury in mice. Toxicol Lett 2010; 192: 387-394.
[78] Metushi IG, Uetrecht J. Isoniazid-induced liver injury and immune response in mice. J Immunotoxicol 2014; 11, 383-392.
[79] Metushi IG, Zhu X, Chen X, Gardam MA, Uetrecht J. Mild isoniazid-induced liver injury in humans is associated with an increase in Th17 cells and T cells producing IL-10. Chem Res Toxicol 2014; 27: 683-689.