1,360
Views
0
CrossRef citations to date
0
Altmetric
Review

Pharmacokinetic and toxicodynamic concepts in idiosyncratic, drug-induced liver injury

, , &
Pages 469-481 | Received 04 Mar 2022, Accepted 11 Aug 2022, Published online: 24 Aug 2022

References

  • Björnsson ES. Drug-induced liver injury: an overview over the most critical compounds. Arch Toxicol. 2015 Mar;89(3):327–334.
  • Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov. 2005 Jun;4(6):489–499.
  • Rao A, Rule JA, Hameed B, et al. Secular trends in severe idiosyncratic drug-induced liver injury in north america: an update from the acute liver failure study group registry. Am J Gastroenterol. 2022 Apr 1;117(4):617–626.
  • Roth RA, Maiuri AR, Ganey PE. Idiosyncratic drug-induced liver injury: is drug-cytokine interaction the linchpin? J Pharmacol Exp Ther. 2017 Feb;360(2):461–470.
  • Hassan A, Fontana RJ. The diagnosis and management of idiosyncratic drug-induced liver injury. Liver Int. 2019 Jan;39(1):31–41.
  • Bunchorntavakul C, Reddy KR. Review article: herbal and dietary supplement hepatotoxicity. Aliment Pharmacol Ther. 2013 Jan;37(1):3–17.
  • Navarro VJ, Khan I, Björnsson E, et al. Liver injury from herbal and dietary supplements. Hepatology. 2017 Jan;65(1):363–373.
  • Koido M, Kawakami E, Fukumura J, et al. Polygenic architecture informs potential vulnerability to drug-induced liver injury. Nat Med. 2020 Oct 1;26(10):1541–1548.
  • Chen M, Borlak J, Tong W. A model to predict severity of drug-induced liver injury in humans. Hepatology. 2016;64(3):931–940.
  • Medina-Caliz I, Robles-Diaz M, Garcia-Muñoz B, et al. Definition and risk factors for chronicity following acute idiosyncratic drug-induced liver injury. J Hepatol. 2016 Sep;65(3):532–542.
  • Korzekwa K. Enzyme kinetics of oxidative metabolism: cytochromes P450. Methods Mol Biol. 2014;1113:149–166.
  • Hart SN, Zhong XB. P450 oxidoreductase: genetic polymorphisms and implications for drug metabolism and toxicity. Expert Opin Drug Metab Toxicol. 2008 Apr;4(4):439–452.
  • Oketch-Rabah HA, Roe AL, Rider CV, et al. United States Pharmacopeia (USP) comprehensive review of the hepatotoxicity of green tea extracts. Toxicol Rep. 2020;7:386–402.
  • Hu J, Webster D, Cao J, et al. The safety of green tea and green tea extract consumption in adults - Results of a systematic review. Regul Toxicol Pharmacol. 2018 Jun;95:412–433.
  • Dostal AM, Samavat H, Bedell S, et al. The safety of green tea extract supplementation in postmenopausal women at risk for breast cancer: results of the Minnesota Green Tea Trial. Food Chem Toxicol. 2015 Sep;83:26–35.
  • Roth RA, Ganey PE. Intrinsic versus idiosyncratic drug-induced hepatotoxicity–two villains or one? J Pharmacol Exp Ther. 2010 Mar;332(3):692–697.
  • Senior JR. What is idiosyncratic hepatotoxicity? What is it not? Hepatology. 2008 Jun;47(6):1813–1815.
  • Marano M, Serafinelli J, Cairoli S, et al. Eltrombopag-induced acute liver failure in a pediatric patient: a pharmacokinetic and pharmacogenetic analysis. Ther Drug Monit. 2018 Aug;40(4):386–388.
  • Bezençon J, Beaudoin JJ, Ito K, et al. Altered expression and function of hepatic transporters in a rodent model of polycystic kidney disease. Drug Metab Dispos. 2019 Aug;47(8):899–906.
  • Beaudoin JJ, Bezençon J, Cao Y, et al. Altered hepatobiliary disposition of tolvaptan and selected tolvaptan metabolites in a rodent model of polycystic kidney disease. Drug Metab Dispos. 2019 Feb;47(2):155–163.
  • Pan G. Roles of hepatic drug transporters in drug disposition and liver toxicity. Adv Exp Med Biol. 2019;1141:293–340.
  • Delgoda R, Westlake AC. Herbal interactions involving cytochrome p450 enzymes: a mini review. Toxicol Rev. 2004;23(4):239–249.
  • Wanwimolruk S, Prachayasittikul V. Cytochrome P450 enzyme mediated herbal drug interactions (Part 1). EXCLI J. 2014;13:347–391.
  • Foti RS, Dickmann LJ, Davis JA, et al. Metabolism and related human risk factors for hepatic damage by usnic acid containing nutritional supplements. Xenobiotica. 2008 Mar;38(3):264–280.
  • Roth RA Jr., Rubin RJ. Role of blood flow in carbon monoxide- and hypoxic hypoxia-induced alterations in hexobarbital metabolism in rats. Drug Metab Dispos. 1976 Sep-Oct;4(5):460–467.
  • Zhou X, Nian Y, Qiao Y, et al. Hypoxia plays a key role in the pharmacokinetic changes of drugs at high altitude. Curr Drug Metab. 2018;19(11):960–969.
  • Hui L, Rong W, Zheng-Ping J, et al. Effects of high altitude exposure on physiology and pharmacokinetics. Curr Drug Metab. 2016;17(6):559–565.
  • Teschke R, Danan G. Idiosyncratic drug induced liver injury, cytochrome P450, metabolic risk factors and lipophilicity: highlights and controversies. Int J Mol Sci. 2021;22(7):3441.
  • Yu K, Geng X, Chen M, et al. High daily dose and being a substrate of cytochrome p450 enzymes are two important predictors of drug-induced liver injury. Drug Metab Dispos. 2014;42(4):744–750.
  • Monks TJ, Anders MW, Dekant W, et al. Glutathione conjugate mediated toxicities. Toxicol Appl Pharmacol. 1990 Oct;106(1):1–19.
  • Boelsterli UA. Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Toxicol Appl Pharmacol. 2003 Nov 1;192(3):307–322.
  • Daly AK. Are polymorphisms in genes relevant to drug disposition predictors of susceptibility to drug-induced liver injury? Pharm Res. 2017 Aug 1;34(8):1564–1569.
  • Yokoi T, Oda S. Models of idiosyncratic drug-induced liver injury. Annu Rev Pharmacol Toxicol. 2021;61(1):247–268.
  • Sasaki E, Matsuo K, Iida A, et al. A novel mouse model for phenytoin-induced liver injury: involvement of immune-related factors and P450-mediated metabolism. Toxicol Sci. 2013 Nov;136(1):250–263.
  • Iida A, Sasaki E, Yano A, et al. Carbamazepine-induced liver injury requires CYP3A-mediated metabolism and glutathione depletion in rats. Drug Metab Dispos. 2015 Jul;43(7):958–968.
  • Chaudhry AS, Urban TJ, Lamba JK, et al. CYP2C9*1B promoter polymorphisms, in linkage with CYP2C19*2, affect phenytoin autoinduction of clearance and maintenance dose. J Pharmacol Exp Ther. 2010 Feb;332(2):599–611.
  • Fleishaker JC, Pearson LK, Peters GR. Phenytoin causes a rapid increase in 6 beta-hydroxycortisol urinary excretion in humans–a putative measure of CYP3A induction. J Pharm Sci. 1995 Mar;84(3):292–294.
  • Oscarson M, Zanger UM, Rifki OF, et al. Transcriptional profiling of genes induced in the livers of patients treated with carbamazepine. Clin Pharmacol Ther. 2006 Nov;80(5):440–456.
  • Waxman DJ, Azaroff L. Phenobarbital induction of cytochrome P-450 gene expression. Biochem J. 1992 Feb 1;281(Pt 3):577–592.
  • Perucca E. Clinically relevant drug interactions with antiepileptic drugs. Br J Clin Pharmacol. 2006 Mar;61(3):246–255.
  • Moon YJ, Wang X, Morris ME. Dietary flavonoids: effects on xenobiotic and carcinogen metabolism. Toxicol In Vitro. 2006 Mar;20(2):187–210.
  • Brewer CT, Chen T. Hepatotoxicity of herbal supplements mediated by modulation of cytochrome P450. Int J Mol Sci. 2017 Nov 8;18(11):2353.
  • Ryu SD, Chung WG. Induction of the procarcinogen-activating CYP1A2 by a herbal dietary supplement in rats and humans. Food Chem Toxicol. 2003 Jun;41(6):861–866.
  • Liu F, Ichihara S, Valentine WM, et al. Increased susceptibility of Nrf2-null mice to 1-bromopropane-induced hepatotoxicity. Toxicol Sci. 2010 Jun;115(2):596–606.
  • Yang M, Zhang H, Tao B, et al. Possible association of HMOX1 and NQO1 polymorphisms with anti-tuberculosis drug-induced liver injury: a matched case-control study. J Clin Pharm Ther. 2019 Aug;44(4):534–542.
  • Huang YS, Su WJ, Huang YH, et al. Genetic polymorphisms of manganese superoxide dismutase,NAD(P)H:quinone oxidoreductase, glutathione S-transferase M1 and T1, and the susceptibility to drug-induced liver injury. J Hepatol. 2007 Jul;47(1):128–134.
  • Junttila MR, Li SP, Westermarck J. Phosphatase-mediated crosstalk between MAPK signaling pathways in the regulation of cell survival. FASEB J. 2008 Apr;22(4):954–965.
  • Maiuri AR, Breier AB, Gora LF, et al. Cytotoxic synergy between cytokines and NSAIDs associated with idiosyncratic hepatotoxicity is driven by mitogen-activated protein kinases. Toxicol Sci. 2015 Aug;146(2):265–280.
  • Xin S, Fang W, Li J, et al. Impact of STAT1 polymorphisms on crizotinib-induced hepatotoxicity in ALK-positive non-small cell lung cancer patients. J Cancer Res Clin Oncol. 2021 Mar;147(3):725–737.
  • Pessayre D, Fromenty B, Berson A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev. 2012 Feb;44(1):34–87.
  • Chan K, Truong D, Shangari N, et al. Drug-induced mitochondrial toxicity. Expert Opin Drug Metab Toxicol. 2005 Dec;1(4):655–669.
  • Luft D, Schmülling RM, Eggstein M. Lactic acidosis in biguanide-treated diabetics: a review of 330 cases. Diabetologia. 1978 Feb;14(2):75–87.
  • Gan SC, Barr J, Arieff AI, et al. Biguanide-associated lactic acidosis. Case report and review of the literature. Arch Internal Med. 1992 Nov;152(11):2333–2336.
  • Dykens JA, Jamieson J, Marroquin L, et al. Biguanide-induced mitochondrial dysfunction yields increased lactate production and cytotoxicity of aerobically-poised HepG2 cells and human hepatocytes in vitro. Toxicol Appl Pharmacol. 2008 Dec 1;233(2):203–210.
  • Will Y, Shields JE, Wallace KB. Drug-induced mitochondrial toxicity in the geriatric population: challenges and future directions. Biology (Basel). 2019 May 11;8(2):32.
  • Gardner K, Hall PA, Chinnery PF, et al. HIV treatment and associated mitochondrial pathology: review of 25 years of in vitro, animal, and human studies. Toxicol Pathol. 2014 Jul;42(5):811–822.
  • Senise JF, Castelo A, Martinez M. Current treatment strategies, complications and considerations for the use of HIV antiretroviral therapy during pregnancy. AIDS Rev. 2011 Oct-Dec;13(4):198–213.
  • Montessori V, Press N, Harris M, et al. Adverse effects of antiretroviral therapy for HIV infection. CMAJ = Journal de l’Association Medicale Canadienne. 2004 Jan 20;170(2):229–238.
  • Montessori V, Harris M, Montaner JS. Hepatotoxicity of nucleoside reverse transcriptase inhibitors. Semin Liver Dis. 2003 May;23(2):167–172.
  • Lewis W, Gonzalez B, Chomyn A, et al. Zidovudine induces molecular, biochemical, and ultrastructural changes in rat skeletal muscle mitochondria. J Clin Invest. 1992 Apr;89(4):1354–1360.
  • Lewis W, Papoian T, Gonzalez B, et al. Mitochondrial ultrastructural and molecular changes induced by zidovudine in rat hearts. Lab Invest. 1991 Aug;65(2):228–236.
  • Ribeiro MP, Santos AE, Custódio JB. Mitochondria: the gateway for tamoxifen-induced liver injury. Toxicology. 2014 Sep 2;323:10–18.
  • Stewart JD, Horvath R, Baruffini E, et al. Polymerase γ gene POLG determines the risk of sodium valproate-induced liver toxicity. Hepatology. 2010 Nov;52(5):1791–1796.
  • Ball AL, Bloch KM, Rainbow L, et al. Assessment of the impact of mitochondrial genotype upon drug-induced mitochondrial dysfunction in platelets derived from healthy volunteers. Arch Toxicol. 2021 Apr;95(4):1335–1347.
  • Kashimshetty R, Desai VG, Kale VM, et al. Underlying mitochondrial dysfunction triggers flutamide-induced oxidative liver injury in a mouse model of idiosyncratic drug toxicity. Toxicol Appl Pharmacol. 2009 Jul 15;238(2):150–159.
  • Shavadia JS, Sharma A, Gu X, et al. Determination of fasiglifam-induced liver toxicity: insights from the data monitoring committee of the fasiglifam clinical trials program. Clin Trial. 2019;16(3):253–262.
  • Li X, Zhong K, Guo Z, et al. Fasiglifam (TAK-875) inhibits hepatobiliary transporters: a possible factor contributing to fasiglifam-induced liver injury. Drug Metab Dispos. 2015 Nov;43(11):1751–1759.
  • Chalasani N, Bonkovsky HL, Fontana R, et al. Features and outcomes of 899 patients with drug-induced liver injury: the DILIN prospective study. Gastroenterology. 2015 Jun;148(7):1340–52 e7.
  • Otieno MA, Snoeys J, Lam W, et al. Fasiglifam (TAK-875): mechanistic investigation and retrospective identification of hazards for drug induced liver injury. Toxicol Sci. 2018 Jun 1;163(2):374–384.
  • Wolenski FS, Zhu AZX, Johnson M, et al. Fasiglifam (TAK-875) alters bile acid homeostasis in rats and dogs: a potential cause of drug induced liver injury. Toxicol Sci. 2017 May 1;157(1):50–61.
  • Li Y, Evers R, Hafey MJ, et al. Use of a bile salt export pump knockdown rat susceptibility model to interrogate mechanism of drug-induced liver toxicity. Toxicol Sci. 2019 Jul 1;170(1):180–198.
  • Hoofnagle JH, Björnsson ES. Drug-induced liver injury - types and phenotypes. N Engl J Med. 2019;381(3):264–273.
  • Matsuo K, Sasaki E, Higuchi S, et al. Involvement of oxidative stress and immune- and inflammation-related factors in azathioprine-induced liver injury. Toxicol Lett. 2014 Jan 13;224(2):215–224.
  • Lee AU, Farrell GC. Mechanism of azathioprine-induced injury to hepatocytes: roles of glutathione depletion and mitochondrial injury. J Hepatol. 2001 Dec;35(6):756–764.
  • Roth RA, Ganey PE. What have we learned from animal models of idiosyncratic, drug-induced liver injury? Expert Opin Drug Metab Toxicol. 2020 Jun;16(6):475–491.
  • Shaw PJ, Ganey PE, Roth RA. Idiosyncratic drug-induced liver injury and the role of inflammatory stress with an emphasis on an animal model of trovafloxacin hepatotoxicity. Toxicol Sci. 2010 Nov;118(1):7–18.
  • Maddox JF, Amuzie CJ, Li M, et al. Bacterial- and viral-induced inflammation increases sensitivity to acetaminophen hepatotoxicity. J Toxicol Environ Health Part A. 2010;73(1):58–73.
  • Schrör K. Aspirin and Reye syndrome: a review of the evidence. Paediatr Drugs. 2007;9(3):195–204.
  • Uetrecht J. Mechanisms of idiosyncratic drug-induced liver injury. Adv Pharmacol. 2019;85:133–163.
  • Kim SH, Naisbitt DJ. Update on advances in research on idiosyncratic drug-induced liver injury. Allergy Asthma Immunol Res. 2016 Jan;8(1):3–11.
  • Daly AK, Day CP. Genetic association studies in drug-induced liver injury. Drug Metab Rev. 2012 Feb;44(1):116–126.
  • Nattrass R, Faulkner L, Vocanson M, et al. Activation of flucloxacillin-specific CD8+ T-cells with the potential to promote hepatocyte cytotoxicity in a mouse model. Toxicol Sci. 2015 Jul;146(1):146–156.
  • Hoofnagle JH, Bonkovsky HL, Phillips EJ, et al. HLA-B*35:01 and green tea-induced liver injury. Hepatology. 2021;73(6):2484–2493.
  • Watkins PB, Kaplowitz N, Slattery JT, et al. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. Jama. 2006 Jul 5;296(1):87–93.
  • Dara L, Liu ZX, Kaplowitz N. Mechanisms of adaptation and progression in idiosyncratic drug induced liver injury, clinical implications. Liver Int. 2016 Feb;36(2):158–165.
  • Mitchell JR, Long MW, Thorgeirsson UP, et al. Acetylation rates and monthly liver function tests during one year of isoniazid preventive therapy. Chest. 1975 Aug;68(2):181–190.
  • Mitchell JR, Zimmerman HJ, Ishak KG, et al. Isoniazid liver injury: clinical spectrum, pathology, and probable pathogenesis. Ann Intern Med. 1976 Feb;84(2):181–192.
  • Metushi I, Uetrecht J, Phillips E. Mechanism of isoniazid-induced hepatotoxicity: then and now. Br J Clin Pharmacol. 2016 Jun;81(6):1030–1036.
  • Watkins PB. Idiosyncratic liver injury: challenges and approaches. Toxicol Pathol. 2005;33(1):1–5.
  • Watkins PB, Zimmerman HJ, Knapp MJ, et al. Hepatotoxic effects of tacrine administration in patients with Alzheimer’s disease. Jama. 1994 Apr 6;271(13):992–998.
  • Danan G, Teschke R. RUCAM in drug and herb induced liver injury: the update. Int J Mol Sci. 2015;17(1):14.
  • Ulrich RG. Idiosyncratic toxicity: a convergence of risk factors. Annu Rev Med. 2007;58:17–34.
  • Li AP. 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. 2002 Nov 10;142(1–2):7–23.