Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-mwx4w Total loading time: 0 Render date: 2024-06-24T16:57:22.332Z Has data issue: false hasContentIssue false

Chapter 22 - Drug-Induced Liver Disease in Children

from Section III - Hepatitis and Immune Disorders

Published online by Cambridge University Press:  19 January 2021

Frederick J. Suchy
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Ronald J. Sokol
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
William F. Balistreri
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Jorge A. Bezerra
Affiliation:
Cincinnati Children’s Hospital Medical Center, Cincinnati
Cara L. Mack
Affiliation:
University of Colorado, Children’s Hospital Colorado, Aurora
Benjamin L. Shneider
Affiliation:
Texas Children’s Hospital, Houston
Get access

Summary

Drug-induced liver disease has been regarded as rare in children. Large surveys have generally failed to detect drug hepatotoxicity as a major problem in children [1], although adverse drug reactions (not necessarily hepatotoxic) are somewhat more frequent in preschool children and in children of any age with cancer. A study examining deaths from adverse drug reactions in children found that approximately one-sixth of such deaths involved acute liver failure, usually associated with antiepileptic or anti-neoplastic drugs [2].

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2021

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Ferrajolo, C, Capuano, A, Verhamme, KM, et al. Drug-induced hepatic injury in children: a case/non-case study of suspected adverse drug reactions in VigiBase. Br J Clin Pharmacol 2010;70:721–8.Google Scholar
Clarkson, A, Choonara, I. Surveillance for fatal suspected adverse drug reactions in the UK. Arch Dis Child 2002;87:462–7.Google Scholar
Squires, RH Jr., Shneider, BL, Bucuvalas, J, et al. Acute liver failure in children: the first 348 patients in the pediatric acute liver failure study group. J Pediatr 2006;148:652–8.Google Scholar
Aithal, GP, Watkins, PB, Andrade, RJ, et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther 2011;89:806–15.CrossRefGoogle ScholarPubMed
Zimmerman, HJ. (1999). Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver, 2nd edn. Philadelphia: Lippincott Williams & Wilkins.Google Scholar
Kaplowitz, N, DeLeve, L. (2013). Drug-Induced Liver Disease, 3rd edn. (p. 776). Waltham, MA: Academic Press.Google Scholar
Ingelman-Sundberg, M. Human drug metabolising cytochrome P450 enzymes: properties and polymorphisms. Naunyn Schmiedebergs Arch Pharmacol 2004;369:89104.CrossRefGoogle ScholarPubMed
McGraw, J, Waller, D. Cytochrome P450 variations in different ethnic populations. Expert Opin Drug Metab Toxicol 2012;8:371–82.CrossRefGoogle ScholarPubMed
Weinshilboum, R. Inheritance and drug response. N Engl J Med 2003;348:529–37.Google Scholar
Cascorbi, I. Genetic basis of toxic reactions to drugs and chemicals. Toxicol Lett 2006;162:1628.Google Scholar
Russmann, S, Jetter, A, Kullak-Ublick, GA. Pharmacogenetics of drug-induced liver injury. Hepatology 2010;52:748–61.CrossRefGoogle ScholarPubMed
Ehmer, U, Kalthoff, S, Fakundiny, B, et al. Gilbert syndrome redefined: a complex genetic haplotype influences the regulation of glucuronidation. Hepatology 2012;55:1912–21.Google Scholar
Burchell, B, Soars, M, Monaghan, G, et al. Drug-mediated toxicity caused by genetic deficiency of UDP-glucuronosyltransferases. Toxicol Lett 2000;112 –13:333–40.Google ScholarPubMed
Pessayre, D, Fromenty, B, Berson, A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev 2012;44:3487.CrossRefGoogle ScholarPubMed
Lucena, MI, Garcia-Martin, E, Andrade, RJ, et al. Mitochondrial superoxide dismutase and glutathione peroxidase in idiosyncratic drug-induced liver injury. Hepatology 2010;52:303–12.Google Scholar
Bessone, F, Dirchwolf, M, Rodil, MA, et al. Review article: drug-induced liver injury in the context of nonalcoholic fatty liver disease – a physiopathological and clinical integrated view. Aliment Pharmacol Ther 2018;48:892913.Google Scholar
He, K, Cai, L, Shi, Q, et al. Inhibition of MDR3 activity in human hepatocytes by drugs associated with liver injury. Chem Res Toxicol 2015;28:1987–90.Google Scholar
Hines, RN, McCarver, DG. The ontogeny of human drug-metabolizing enzymes: phase I oxidative enzymes. J Pharmacol Exp Ther 2002;300:355–60.Google Scholar
Kearns, GL, Abdel-Rahman, SM, Alander, SW, et al. Developmental pharmacology–drug disposition, action, and therapy in infants and children. N Engl J Med 2003;349:1157–67.CrossRefGoogle ScholarPubMed
Tateishi, T, Nakura, H, Asoh, M, et al. A comparison of hepatic cytochrome P450 protein expression between infancy and postinfancy. Life Sci 1997;61:2567–74.Google Scholar
Aranda, JV, Collinge, JM, Zinman, R, et al. Maturation of caffeine elimination in infancy. Arch Dis Child 1979;54:946–9.Google Scholar
Treluyer, JM, Gueret, G, Cheron, G, et al. Developmental expression of CYP2C and CYP2C-dependent activities in the human liver: in-vivo/in-vitro correlation and inducibility. Pharmacogenetics 1997;7:441–52.Google Scholar
Lang, C, Meier, Y, Stieger, B, et al. Mutations and polymorphisms in the bile salt export pump and the multidrug resistance protein 3 associated with drug-induced liver injury. Pharmacogenet Genomics 2007;17:4760.CrossRefGoogle ScholarPubMed
Molleston, JP, Fontana, RJ, Lopez, MJ, et al. Characteristics of idiosyncratic drug-induced liver injury in children: results from the DILIN prospective study. J Pediatr Gastroenterol Nutr 2011;53:182–9.Google Scholar
Walsh, SA, Creamer, D. Drug reaction with eosinophilia and systemic symptoms (DRESS): a clinical update and review of current thinking. Clin Exp Dermatol 2011;36:611.Google Scholar
Cacoub, P, Musette, P, Descamps, V, et al. The DRESS syndrome: a literature review. Am J Med 2011;124:588–97.CrossRefGoogle ScholarPubMed
Devarbhavi, H, Karanth, D, Prasanna, KS, et al. Drug-induced liver injury with hypersensitivity features has a better outcome: a single-center experience of 39 children and adolescents. Hepatology 2011;54:1344–50.CrossRefGoogle Scholar
Gudnason, HO, Bjornsson, HK, Gardarsdottir, M, et al. Secondary sclerosing cholangitis in patients with drug-induced liver injury. Dig Liver Dis 2015;47:502–7.CrossRefGoogle ScholarPubMed
Jaeschke, H, Gores, GJ, Cederbaum, AI, et al. Mechanisms of hepatotoxicity. Toxicol Sci 2002;65:166–76.Google Scholar
James, LP, Farrar, HC, Darville, TL, et al. Elevation of serum interleukin 8 levels in acetaminophen overdose in children and adolescents. Clin Pharmacol Ther 2001;70:280–6.Google Scholar
Ju, C, Reilly, TP, Bourdi, M, et al. Protective role of Kupffer cells in acetaminophen-induced hepatic injury in mice. Chem Res Toxicol 2002;15:1504–13.Google Scholar
Aithal, GP, Ramsay, L, Daly, AK, et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology 2004;39:1430–40.CrossRefGoogle ScholarPubMed
Kaddurah-Daouk, R, Weinshilboum, R, Pharmacometabolomics Research. Metabolomic signatures for drug response phenotypes: pharmacometabolomics enables precision medicine. Clin Pharmacol Ther 2015;98:71–5.Google Scholar
Enright, EF, Gahan, CG, Joyce, SA, et al. The impact of the gut microbiota on drug metabolism and clinical outcome. Yale J Biol Med 2016;89:375–82.Google Scholar
Massart, J, Begriche, K, Moreau, C, et al. Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity. J Clin Transl Res 2017;3:212–32.Google Scholar
DiPaola, F, Molleston, JP, Gu, J, et al. Antimicrobials and antiepileptics are the leading causes of idiosyncratic drug-induced liver injury in American children. J Pediatr Gastroenterol Nutr 2019;69:152–9.Google Scholar
Mahadevan, SB, McKiernan, PJ, Davies, P, et al. Paracetamol induced hepatotoxicity. Arch Dis Child 2006;91:598603.CrossRefGoogle ScholarPubMed
Makin, AJ, Wendon, J, Williams, R. A 7-year experience of severe acetaminophen-induced hepatotoxicity (1987–1993). Gastroenterology 1995;109:1907–16.CrossRefGoogle ScholarPubMed
Smilkstein, MJ, Knapp, GL, Kulig, KW, et al. Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. N Engl J Med 1988;319:1557–62.Google Scholar
Harrison, PM, Keays, R, Bray, GP, et al. Improved outcome of paracetamol-induced fulminant hepatic failure by late administration of acetylcysteine. Lancet 1990;335:1572–3.Google Scholar
Keays, R, Harrison, PM, Wendon, JA, et al. Intravenous acetylcysteine in paracetamol induced fulminant hepatic failure: a prospective controlled trial. BMJ 1991;303:1026–9.CrossRefGoogle ScholarPubMed
Yarema, MC, Johnson, DW, Berlin, RJ, et al. Comparison of the 20-hour intravenous and 72-hour oral acetylcysteine protocols for the treatment of acute acetaminophen poisoning. Ann Emerg Med 2009;54:606–14.CrossRefGoogle ScholarPubMed
Gosselin, S, Juurlink, DN, Kielstein, JT, et al. Extracorporeal treatment for acetaminophen poisoning: recommendations from the EXTRIP workgroup. Clin Toxicol (Phila) 2014;52:856–67.Google Scholar
James, LP, Wells, E, Beard, RH, et al. Predictors of outcome after acetaminophen poisoning in children and adolescents. J Pediatr 2002;140:522–6.Google Scholar
Heubi, JE, Barbacci, MB, Zimmerman, HJ. Therapeutic misadventures with acetaminophen: hepatoxicity after multiple doses in children. J Pediatr 1998;132:22–7.Google Scholar
Leonis, MA, Alonso, EM, Im, K, et al. Chronic acetaminophen exposure in pediatric acute liver failure. Pediatrics 2013;131:e740–6.Google Scholar
Rajanayagam, J, Bishop, JR, Lewindon, PJ, et al. Paracetamol-associated acute liver failure in Australian and New Zealand children: high rate of medication errors. Arch Dis Child 2015;100:7780.Google Scholar
Acheampong, P, Thomas, SH. Determinants of hepatotoxicity after repeated supratherapeutic paracetamol ingestion: systematic review of reported cases. Br J Clin Pharmacol 2016;82:923–31.Google Scholar
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;296:8793.Google Scholar
Savino, F, Lupica, MM, Tarasco, V, et al. Fulminant hepatitis after 10 days of acetaminophen treatment at recommended dosage in an infant. Pediatrics 2011;127:e494–7.Google Scholar
Iorio, ML, Cheerharan, M, Kaufman, SS, et al. Acute liver failure following cleft palate repair: a case of therapeutic acetaminophen toxicity. Cleft Palate Craniofac J 2013;50:747–50.Google Scholar
Webster, PA, Roberts, DW, Benson, RW, et al. Acetaminophen toxicity in children: diagnostic confirmation using a specific antigenic biomarker. J Clin Pharmacol 1996;36:397402.Google Scholar
Bhattacharyya, S, Yan, K, Pence, L, et al. Targeted liquid chromatography-mass spectrometry analysis of serum acylcarnitines in acetaminophen toxicity in children. Biomark Med 2014;8:147–59.Google Scholar
Yang, X, Salminen, WF, Shi, Q, et al. Potential of extracellular microRNAs as biomarkers of acetaminophen toxicity in children. Toxicol Appl Pharmacol 2015;284:180–7.Google Scholar
Jaeschke, H, Knight, TR, Bajt, ML. The role of oxidant stress and reactive nitrogen species in acetaminophen hepatotoxicity. Toxicol Lett 2003;144:279–88.Google Scholar
Fannin, RD, Russo, M, O’Connell, TM, et al. Acetaminophen dosing of humans results in blood transcriptome and metabolome changes consistent with impaired oxidative phosphorylation. Hepatology 2010;51:227–36.Google Scholar
Goldring, CE, Kitteringham, NR, Elsby, R, et al. Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice. Hepatology 2004;39:1267–76.Google Scholar
Liu, HH, Lu, P, Guo, Y, et al. An integrative genomic analysis identifies Bhmt2 as a diet-dependent genetic factor protecting against acetaminophen-induced liver toxicity. Genome Res 2010;20:2835.Google Scholar
Harrill, AH, Watkins, PB, Su, S, et al. Mouse population-guided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans. Genome Res 2009;19:1507–15.Google Scholar
Zamora, R, Barclay, D, Yin, J, et al. HMGB1 is a central driver of dynamic pro-inflammatory networks in pediatric acute liver failure induced by acetaminophen. Sci Rep 2019;9:5971.Google Scholar
Bourdi, M, Masubuchi, Y, Reilly, TP, et al. Protection against acetaminophen-induced liver injury and lethality by interleukin 10: role of inducible nitric oxide synthase. Hepatology 2002;35:289–98.Google Scholar
Connolly, MK, Ayo, D, Malhotra, A, et al. Dendritic cell depletion exacerbates acetaminophen hepatotoxicity. Hepatology 2011;54:959–68.CrossRefGoogle ScholarPubMed
Peterson, RG, Rumack, BH. Age as a variable in acetaminophen overdose. Arch Intern Med 1981;141:390–3.Google Scholar
Rumack, BH. Acetaminophen overdose in young children. Treatment and effects of alcohol and other additional ingestants in 417 cases. Am J Dis Child 1984;138:428–33.Google Scholar
Peterson, RG, Rumack, BH. Pharmacokinetics of acetaminophen in children. Pediatrics 1978;62:877–9.CrossRefGoogle ScholarPubMed
Hickson, GB, Altemeier, WA, Martin, ED, et al. Parental administration of chemical agents: a cause of apparent life threatening events. J Pediatr 1989;83:772–6.Google Scholar
Pacifici, GM, Allegaert, K. Clinical pharmacology of paracetamol in neonates: a review. Curr Ther Res Clin Exp 2015;77:2430.Google Scholar
Cook, SF, Stockmann, C, Samiee-Zafarghandy, S, et al. Neonatal maturation of paracetamol (acetaminophen) glucuronidation, sulfation, and oxidation based on a parent-metabolite population pharmacokinetic model. Clin Pharmacokinet 2016;55:13951411.Google Scholar
Flint, RB, Roofthooft, DW, van Rongen, A, et al. Exposure to acetaminophen and all its metabolites upon 10, 15, and 20 mg/kg intravenous acetaminophen in very-preterm infants. Pediatr Res 2017;82:678–84.Google Scholar
Walls, L, Baker, CF, Sarkar, S. Acetaminophen-induced hepatic failure with encephalopathy in a newborn. J Perinatol 2007;27:133–5.Google Scholar
Ogilvie, JD, Rieder, MJ, Lim, R. Acetaminophen overdose in children. CMAJ 2012;184:1492–6.Google Scholar
Palmer, GM, Atkins, M, Anderson, BJ, et al. I.V. acetaminophen pharmacokinetics in neonates after multiple doses. Br J Anaesth 2008;101:523–30.Google Scholar
Rumack, BH. Acetaminophen misconceptions. Hepatology 2004;40:1015.Google Scholar
Al-Sinani, S, Al-Rawas, A, Dhawan, A. Mercury as a cause of fulminant hepatic failure in a child: case report and literature review. Clin Res Hepatol Gastroenterol 2011;35:580–2.CrossRefGoogle Scholar
Zimmerman, HJ, Maddrey, WC. Acetaminophen (paracetamol) hepatotoxicity with regular intake of alcohol: analysis of instances of therapeutic misadventure. Hepatology 1995;22:767–73.Google Scholar
Bruun, LS, Elkjaer, S, Bitsch-Larsen, D, et al. Hepatic failure in a child after acetaminophen and sevoflurane exposure. Anesth Analg 2001;92:1446–8.Google Scholar
Ceschi, A, Hofer, KE, Rauber-Luthy, C, et al. Paracetamol orodispersible tablets: a risk for severe poisoning in children? Eur J Clin Pharmacol 2011;67:97–9.Google Scholar
Dart, RC, Rumack, BH. Intravenous acetaminophen in the United States: iatrogenic dosing errors. Pediatrics 2012;129:349–53.Google Scholar
Berling, I, Anscombe, M, Isbister, GK. Intravenous paracetamol toxicity in a malnourished child. Clin Toxicol (Phila) 2012;50:74–6.Google Scholar
Yagupsky, P, Gazala, E, Sofer, S. Fatal hepatic failure and encephalopathy associated with amiodarone therapy. J Pediatr 1985;107:967–70.Google Scholar
Floyd, J, Mirza, I, Sachs, B, et al. Hepatotoxicity of chemotherapy. Semin Oncol 2006;33:5067.Google Scholar
Pratibha, R, Sameer, R, Rataboli, PV, et al. Enzymatic studies of cisplatin induced oxidative stress in hepatic tissue of rats. Eur J Pharmacol 2006;532:290–3.Google Scholar
Lu, Y, Cederbaum, AI. Cisplatin-induced hepatotoxicity is enhanced by elevated expression of cytochrome P450 2E1. Toxicol Sci 2006;89:515–23.Google Scholar
Hruban, RH, Sternberg, SS, Meyers, P, et al. Fatal thrombocytopenia and liver failure associated with carboplatin therapy. Cancer Invest 1991;9:263–8.Google Scholar
Honjo, I, Suou, T, Hirayama, C. Hepatotoxicity of cyclophosphamide in man: pharmacokinetic analysis. Res Commun Chem Pathol Pharmacol 1988;61:149–65.Google Scholar
Berkovitch, M, Matsui, D, Zipursky, A, et al. Hepatotoxicity of 6-mercaptopurine in childhood acute lymphocytic leukemia: pharmacokinetic characteristics. Med Pediatr Oncol 1996;26:85–9.Google Scholar
Hazar, V, Kutluk, T, Akyuz, C, et al. Veno-occlusive disease-like hepatotoxicity in two children receiving chemotherapy for Wilms’ tumor and clear cell sarcoma of kidney. Pediatr Hematol Oncol 1998;15:85–9.Google Scholar
Shi, Q, Yang, X, Greenhaw, JJ, et al. Drug-induced liver injury in children: clinical observations, animal models, and regulatory status. Int J Toxicol 2017;36:365–79.CrossRefGoogle ScholarPubMed
Pratt, CB, Johnson, WW. Duration and severity of fatty metamorphosis of the liver following L-asparaginase therapy. Cancer 1971;28:361–4.Google Scholar
Sahoo, S, Hart, J. Histopathological features of L-asparaginase-induced liver disease. Semin Liver Dis 2003;23:295–9.Google Scholar
Hijiya, N, van der Sluis, IM. Asparaginase-associated toxicity in children with acute lymphoblastic leukemia. Leuk Lymphoma 2016;57:748–57.Google Scholar
Topley, J, Benson, J, Squier, MV, et al. Hepatotoxicity in the treatment of acute lymphoblastic leukemia. Med Pediatr Oncol 1979;7:393–9.Google Scholar
Stoneham, S, Lennard, L, Coen, P, et al. Veno-occlusive disease in patients receiving thiopurines during maintenance therapy for childhood acute lymphoblastic leukaemia. Br J Haematol 2003;123:100–2.Google Scholar
Broxson, EH, Dole, M, Wong, R, et al. Portal hypertension develops in a subset of children with standard risk acute lymphoblastic leukemia treated with oral 6-thioguanine during maintenance therapy. Pediatr Blood Cancer 2005;44:226–31.Google Scholar
Rawat, D, Gillett, PM, Devadason, D, et al. Long-term follow-up of children with 6-thioguanine-related chronic hepatotoxicity following treatment for acute lymphoblastic leukaemia. J Pediatr Gastroenterol Nutr 2011;53:478–9.Google Scholar
Geller, SA, Dubinsky, MC, Poordad, FF, et al. Early hepatic nodular hyperplasia and submicroscopic fibrosis associated with 6-thioguanine therapy in inflammatory bowel disease. Am J Surg Pathol 2004;28:1204–11.Google Scholar
DeLeve, LD, Shulman, HM, McDonald, GB. Toxic injury to hepatic sinusoids: sinusoidal obstruction syndrome (veno-occlusive disease). Semin Liver Dis 2002;22:2742.CrossRefGoogle ScholarPubMed
D’Antiga, L, Baker, A, Pritchard, J, et al. Veno-occlusive disease with multi-organ involvement following actinomycin-D. Eur J Cancer 2001;37:1141–8.Google Scholar
Barker, CC, Anderson, RA, Sauve, RS, et al. GI complications in pediatric patients post-BMT. Bone Marrow Transplant 2005;36:51–8.Google Scholar
Sulis, ML, Bessmertny, O, Granowetter, L, et al. Veno-occlusive disease in pediatric patients receiving actinomycin D and vincristine only for the treatment of rhabdomyosarcoma. J Pediatr Hematol Oncol 2004;26:843–6.Google Scholar
Elli, M, Pinarli, FG, Dagdemir, A, et al. Veno-occlusive disease of the liver in a child after chemotherapy for brain tumor. Pediatr Blood Cancer 2006;46:521–3.Google Scholar
McDonald, GB, Sharma, P, Matthews, DE, et al. The clinical course of 53 patients with veno-occlusive disease of the liver after marrow transplantation. Transplantation 1985;39:603–8.CrossRefGoogle Scholar
McDonald, GB, Slattery, JT, Bouvier, ME, et al. Cyclophosphamide metabolism, liver toxicity, and mortality following hematopoietic stem cell transplantation. Blood 2003;101:2043–8.Google Scholar
El-Sayed, MH, El-Haddad, A, Fahmy, OA, et al. Liver disease is a major cause of mortality following allogeneic bone-marrow transplantation. Eur J Gastroenterol Hepatol 2004;16:1347–54.Google Scholar
Srivastava, A, Poonkuzhali, B, Shaji, RV, et al. Glutathione S-transferase M1 polymorphism: a risk factor for hepatic venoocclusive disease in bone marrow transplantation. Blood 2004;104:1574–7.Google Scholar
Reiss, U, Cowan, M, McMillan, A, et al. Hepatic venoocclusive disease in blood and bone marrow transplantation in children and young adults: incidence, risk factors, and outcome in a cohort of 241 patients. J Pediatr Hematol Oncol 2002;24:746–50.Google Scholar
Ravikumara, M, Hill, FG, Wilson, DC, et al. 6-Thioguanine-related chronic hepatotoxicity and variceal haemorrhage in children treated for acute lymphoblastic leukaemia–a dual-centre experience. J Pediatr Gastroenterol Nutr 2006;42:535–8.Google Scholar
Corbacioglu, S, Greil, J, Peters, C, et al. Defibrotide in the treatment of children with veno-occlusive disease (VOD): a retrospective multicentre study demonstrates therapeutic efficacy upon early intervention. Bone Marrow Transplant 2004;33:189–95.Google Scholar
Shin-Nakai, N, Ishida, H, Yoshihara, T, et al. Control of hepatic veno-occlusive disease with an antithrombin-III concentrate-based therapy. Pediatr Int 2006;48:85–7.Google Scholar
Ringden, O, Remberger, M, Lehmann, S, et al. N-acetylcysteine for hepatic veno-occlusive disease after allogeneic stem cell transplantation. Bone Marrow Transplant 2000;25:993–6.Google Scholar
Lee, AC, Goh, PY. Dactinomycin-induced hepatic sinusoidal obstruction syndrome responding to treatment with N-acetylcysteine. J Cancer 2011;2:527–31.Google Scholar
DeLeve, LD. Dacarbazine toxicity in murine liver cells: a model of hepatic endothelial injury and glutathione defense. J Pharmacol Exp Ther 1994;268:1261–70.Google Scholar
DeLeve, LD. Cellular target of cyclophosphamide toxicity in the murine liver: role of glutathione and site of metabolic activation. Hepatology 1996;24:830–7.Google Scholar
DeLeve, LD, Wang, X, Kanel, GC, et al. Decreased hepatic nitric oxide production contributes to the development of rat sinusoidal obstruction syndrome. Hepatology 2003;38:900–8.Google Scholar
Iguchi, A, Kobayashi, R, Yoshida, M, et al. Vascular endothelial growth factor (VEGF) is one of the cytokines causative and predictive of hepatic veno-occlusive disease (VOD) in stem cell transplantation. Bone Marrow Transplant 2001;27:1173–80.Google Scholar
Suzman, DL, Pelosof, L, Rosenberg, A, et al. Hepatotoxicity of immune checkpoint inhibitors: an evolving picture of risk associated with a vital class of immunotherapy agents. Liver Int 2018;38:976–87.Google Scholar
Chen, TC, Ng, KF, Jeng, LB, et al. Aspirin-related hepatotoxicity in a child after liver transplant. Dig Dis Sci 2001;46:486–8.Google Scholar
Hamdan, JA, Manasra, K, Ahmed, M. Salicylate-induced hepatitis in rheumatic fever. Am J Dis Child 1985;139:453–5.Google Scholar
Dinakaran, D, Bristow, E, Armanious, H, et al. Co-ingestion of willow bark tea and acetaminophen associated with fatal infantile fulminant liver failure. Pediatr Int 2017;59:743–5.Google Scholar
Raza, A, Vierling, J, Hussain, KB. Genetics of drug-induced hepatotoxicity toxicity in Gilbert’s syndrome. Am J Gastroenterol 2013;108:1936–7.Google Scholar
Dinh, JC, Pearce, RE, Van Haandel, L, et al. Characterization of atomoxetine biotransformation and implications for development of PBPK models for dose individualization in children. Drug Metab Dispos 2016;44:1070–9.Google Scholar
Stojanovski, SD, Casavant, MJ, Mousa, HM, et al. Atomoxetine-induced hepatitis in a child. Clin Toxicol (Phila) 2007;45:51–5.Google Scholar
Erdogan, A, Ozcay, F, Piskin, E, et al. Idiosyncratic liver failure probably associated with atomoxetine: a case report. J Child Adolesc Psychopharmacol 2011;21:295–7.Google Scholar
Lim, JR, Faught, PR, Chalasani, NP, et al. Severe liver injury after initiating therapy with atomoxetine in two children. J Pediatr 2006;148:831–4.Google Scholar
Brown, JT, Abdel-Rahman, SM, van Haandel, L, et al. Single dose, CYP2D6 genotype-stratified pharmacokinetic study of atomoxetine in children with ADHD. Clin Pharmacol Ther 2016;99:642–50.Google Scholar
DePinho, RA, Goldberg, CS, Lefkowitch, JH. Azathioprine and the liver. Evidence favoring idiosyncratic, mixed cholestatic-hepatocellular injury in humans. Gastroenterology 1984;86:162–5.Google Scholar
Sterneck, M, Wiesner, R, Ascher, N, et al. Azathioprine hepatotoxicity after liver transplantation. Hepatology 1991;14:806–10.Google Scholar
Seiderer, J, Zech, CJ, Diebold, J, et al. Nodular regenerative hyperplasia: a reversible entity associated with azathioprine therapy. Eur J Gastroenterol Hepatol 2006;18:553–5.Google Scholar
Bjornsson, ES, Gu, J, Kleiner, DE, et al. Azathioprine and 6-mercaptopurine-induced liver injury: clinical features and outcomes. J Clin Gastroenterol 2017;51:63–9.Google Scholar
Yang, SK, Hong, M, Baek, J, et al. A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 2014;46:1017–20.Google Scholar
Wang, CH, Chi, MH, Lin, JY, et al. NUDT15 polymorphism identified in a patient with azathioprine hypersensitivity syndrome presenting as erythema nodosum and hepatotoxicity. Br J Dermatol 2019;181:631–2.Google Scholar
Lennard, L. The clinical pharmacology of 6-mercaptopurine. Eur J Clin Pharmacol 1992;43:329–39.Google Scholar
Adam de Beaumais, T, Fakhoury, M, Medard, Y, et al. Determinants of mercaptopurine toxicity in paediatric acute lymphoblastic leukemia maintenance therapy. Br J Clin Pharmacol 2011;71:575–84.Google Scholar
Rosh, JR, Gross, T, Mamula, P, et al. Hepatosplenic T-cell lymphoma in adolescents and young adults with Crohn’s disease: a cautionary tale? Inflamm Bowel Dis 2007;13:1024–30.Google Scholar
Ochenrider, MG, Patterson, DJ, Aboulafia, DM. Hepatosplenic T-cell lymphoma in a young man with Crohn’s disease: case report and literature review. Clin Lymphoma Myeloma Leuk 2010;10:144–8.Google Scholar
Deepak, P, Sifuentes, H, Sherid, M, et al. T-cell non-Hodgkin’s lymphomas reported to the FDA AERS with tumor necrosis factor-alpha (TNF-alpha) inhibitors: results of the REFURBISH study. Am J Gastroenterol 2013;108:99105.Google Scholar
Bedan, M, Grimm, D, Wehland, M, et al. A focus on macitentan in the treatment of pulmonary arterial hypertension. Basic Clin Pharmacol Toxicol 2018;123:103–13.Google Scholar
Fattinger, K, Funk, C, Pantze, M, et al. The endothelin antagonist bosentan inhibits the canalicular bile salt export pump: a potential mechanism for hepatic adverse reactions. Clin Pharmacol Ther 2001;69:223–31.CrossRefGoogle ScholarPubMed
Wei, A, Gu, Z, Li, J, et al. Clinical adverse effects of endothelin receptor antagonists: insights from the meta-analysis of 4894 patients from 24 randomized double-blind placebo-controlled clinical trials. J Am Heart Assoc 2016;5(11):e003896.Google Scholar
Eriksson, C, Gustavsson, A, Kronvall, T, et al. Hepatotoxicity by bosentan in a patient with portopulmonary hypertension: a case-report and review of the literature. J Gastrointestin Liver Dis 2011;20:7780.Google Scholar
Beghetti, M, Hoeper, MM, Kiely, DG, et al. Safety experience with bosentan in 146 children 2–11 years old with pulmonary arterial hypertension: results from the European Postmarketing Surveillance Program. Pediatr Res 2008;64:200–4.Google Scholar
Berger, RM, Haworth, SG, Bonnet, D, et al. FUTURE-2: results from an open-label, long-term safety and tolerability extension study using the pediatric formulation of bosentan in pulmonary arterial hypertension. Int J Cardiol 2016;202:52–8.Google Scholar
Shear, NH, Spielberg, SP. Anticonvulsant hypersensitivity syndrome. In vitro assessment of risk. J Clin Invest 1988;82:1826–32.Google Scholar
Zucker, P, Daum, F, Cohen, MI. Fatal carbamazepine hepatitis. J Pediatr 1977;91:667–8.Google Scholar
Smith, DW, Cullity, GJ, Silberstein, EP. Fatal hepatic necrosis associated with multiple anticonvulsant therapy. Aust N Z J Med 1988;18:575–81.Google Scholar
Hadzic, N, Portmann, B, Davies, ET, et al. Acute liver failure induced by carbamazepine. Arch Dis Child 1990;65:315–17.Google Scholar
Morales-Diaz, M, Pinilla-Roa, E, Ruiz, I. Suspected carbamazepine-induced hepatotoxicity. Pharmacotherapy 1999;19:252–5.Google Scholar
Sierra, NM, Garcia, B, Marco, J, et al. Cross hypersensitivity syndrome between phenytoin and carbamazepine. Pharm World Sci 2005;27:170–4.Google Scholar
Aouam, K, Ben Romdhane, F, Loussaief, C, et al. Hypersensitivity syndrome induced by anticonvulsants: possible cross-reactivity between carbamazepine and lamotrigine. J Clin Pharmacol 2009;49:1488–91.Google Scholar
Yip, VLM, Meng, X, Maggs, JL, et al. Mass spectrometric characterization of circulating covalent protein adducts derived from epoxide metabolites of carbamazepine in patients. Chem Res Toxicol 2017;30:1419–35.Google Scholar
Ju, C, Uetrecht, J. Detection of 2-hydroxyiminostilbene in the urine of patients taking carbamazepine and its oxidation to a reactive iminequinone intermediate. J Pharmacol Exp Ther 1999;288:51–6.Google Scholar
McCormack, M, Alfirevic, A, Bourgeois, S, et al. HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans. N Engl J Med 2011;364:1134–43.Google Scholar
Phillips, EJ, Sukasem, C, Whirl-Carrillo, M, et al. Clinical Pharmacogenetics Implementation Consortium guideline for HLA genotype and use of carbamazepine and oxcarbazepine: 2017 update. Clin Pharmacol Ther 2018;103:574–81.Google Scholar
D’Orazio, JL. Oxcarbazepine-induced Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS). Clin Toxicol (Phila) 2008;46:1093–4.Google Scholar
Bosdure, E, Cano, A, Roquelaure, B, et al. Oxcarbazepine and DRESS syndrome: a paediatric cause of acute liver failure. Arch Pediatr 2004;11:1073–7.Google Scholar
Wanless, IR, Dore, S, Gopinath, G, et al. Histopathology of cocaine hepatotoxicity. Report of four cases. Gastroenterology 1990;98:497501.Google Scholar
Kassianides, C, Nussenblatt, R, Palestine, AG, et al. Liver injury from cyclosporine A. Dig Dis Sci 1990;35:693–7.Google Scholar
Brauer, RB, Heidecke, CD, Nathrath, W, et al. Liver transplantation for the treatment of fulminant hepatic failure induced by the ingestion of ecstasy. Transpl Int 1997;10:229–33.Google Scholar
Andreu, V, Mas, A, Bruguera, M, et al. Ecstasy: a common cause of severe acute hepatotoxicity. J Hepatol 1998;29:394–7.Google Scholar
Greene, SL, Dargan, PI, O’Connor, N, et al. Multiple toxicity from 3,4-methylenedioxymethamphetamine (“Ecstasy”). Am J Emerg Med 2003;21:121–4.Google Scholar
Smith, ID, Simpson, KJ, Garden, OJ, et al. Non-paracetamol drug-induced fulminant hepatic failure among adults in Scotland. Eur J Gastroenterol Hepatol 2005;17:161–7.Google Scholar
Antolino-Lobo, I, Meulenbelt, J, van den Berg, M, et al. A mechanistic insight into 3,4-methylenedioxymethamphetamine (“Ecstasy”)-mediated hepatotoxicity. Vet Q 2011;31:193205.Google Scholar
Ellis, AJ, Wendon, JA, Portmann, B, et al. Acute liver damage and ecstasy ingestion. Gut 1996;38:454–8.Google Scholar
Munoz, P, Drobinska, A, Bianchi, L, et al. Acute giant cell hepatitis in a 17-year-old man. Schweiz Rundsch Med Prax 2004;93:2109–12.Google Scholar
Duffy, MR, Swart, M. Severe Ecstasy poisoning in a toddler. Anaesthesia 2006;61:498501.Google Scholar
Yin, S. Adolescents and drug abuse: 21st century synthetic substances. Clin Pedaitr Emerg Med 2019;20:1724.Google Scholar
Hood, B, Nowicki, MJ. Eosinophilic hepatitis in an adolescent during lisdexamfetamine dimesylate treatment for ADHD. Pediatrics 2010;125:e1510–13.Google Scholar
Zafrani, ES, Ishak, KG, Rudzki, C. Cholestatic and hepatocellular injury associated with erythromycin esters. Report of nine cases. Dig Dis Sci 1979;24:385–96.Google Scholar
Phillips, KG. Hepatotoxicity of erythromycin ethylsuccinate in a child. Can Med Assoc J 1983;129:411–12.Google Scholar
Principi, N, Esposito, S. Comparative tolerability of erythromycin and newer macrolide antibacterials in paediatric patients. Drug Saf 1999;20:2541.Google Scholar
Karthik, SV, Casson, D. Erythromycin-associated cholestatic hepatitis and liver dysfunction in children: the British experience. J Clin Gastroenterol 2005;39:743–4.Google Scholar
Woodhead, JL, Yang, K, Oldach, D, et al. Analyzing the mechanisms behind macrolide antibiotic-induced liver injury using quantitative systems toxicology modeling. Pharm Res 2019;36:48.Google Scholar
Martinez, MA, Vuppalanchi, R, Fontana, RJ, et al. Clinical and histologic features of azithromycin-induced liver injury. Clin Gastroenterol Hepatol 2015;13:369–76 e363.Google Scholar
Giannattasio, A, D’Ambrosi, M, Volpicelli, M, et al. Steroid therapy for a case of severe drug-induced cholestasis. Ann Pharmacother 2006;40:1196–9.Google Scholar
Fox, JC, Szyjkowski, RS, Sanderson, SO, et al. Progressive cholestatic liver disease associated with clarithromycin treatment. J Clin Pharmacol 2002;42:676–80.Google Scholar
Brinker, AD, Wassel, RT, Lyndly, J, et al. Telithromycin-associated hepatotoxicity: clinical spectrum and causality assessment of 42 cases. Hepatology 2009;49:250–7.Google Scholar
Neuberger, J, Nunnerley, HB, Davis, M, et al. Oral-contraceptive-associated liver tumours: occurrence of malignancy and difficulties in diagnosis. Lancet 1980;i:273–6.Google Scholar
Nault, JC, Bioulac-Sage, P, Zucman-Rossi, J. Hepatocellular benign tumors: from molecular classification to personalized clinical care. Gastroenterology 2013;144:888902.Google Scholar
Jeannot, E, Poussin, K, Chiche, L, et al. Association of CYP1B1 germ line mutations with hepatocyte nuclear factor 1alpha-mutated hepatocellular adenoma. Cancer Res 2007;67:2611–16.CrossRefGoogle ScholarPubMed
Pellock, JM, Faught, E, Leppik, IE, et al. Felbamate: consensus of current clinical experience. Epilepsy Res 2006;71:89101.Google Scholar
Popovic, M, Nierkens, S, Pieters, R, et al. Investigating the role of 2-phenylpropenal in felbamate-induced idiosyncratic drug reactions. Chem Res Toxicol 2004;17:1568–76.Google Scholar
Gaertner, I, Altendorf, K, Batra, A, et al. Relevance of liver enzyme elevations with four different neuroleptics: a retrospective review of 7,263 treatment courses. J Clin Psychopharmacol 2001;21:215–22.Google Scholar
Lo, SK, Wendon, J, Mieli-Vergani, G, et al. Halothane-induced acute liver failure: continuing occurrence and use of liver transplantation. Eur J Gastroenterol Hepatol 1998;10:635–9.Google Scholar
Kenna, JG, Newberger, J, Mieli-Vergani, G, et al. Halothane hepatitis in children. Br Med J 1987;294:1209–11.Google Scholar
Hassall, E, Israel, DM, Gunasekaran, T, et al. Halothane hepatitis in children. J Pediatr Gastroenterol Nutr 1990;11:553–7.Google Scholar
Wark, HJ. Postoperative jaundice in children. The influence of halothane. Anaesthesia 1983;38:237–42.Google Scholar
Warner, LO, Beach, TP, Gariss, JP, et al. Halothane and children: the first quarter century. Anesth Analg 1984;63:838–40.Google Scholar
Psacharopoulos, HJ, Mowat, AP, Davies, M, et al. Fulminant hepatic failure in childhood: an analysis of 31 cases. Arch Dis Child 1980;55:252–8.Google Scholar
Farrell, GC. Mechanism of halothane-induced liver injury: is it immune or metabolic idiosyncrasy? J Gastroenterol Hepatol 1988;3:465–82.Google Scholar
Pohl, LR, Satoh, H, Christ, DD, et al. The immunologic and metabolic basis of drug hypersensitivities. Annu Rev Pharmacol Toxicol 1988;28:367–87.Google Scholar
Kenna, JG, Satoh, H, Christ, DD, et al. Metabolic basis for a drug hypersensitivity: antibodies in sera from patients with halothane hepatitis recognize liver neoantigens that contain the trifluoroacetyl group derived from halothane. J Pharmacol Exp Ther 1988;245:1103–9.Google Scholar
Kenna, JG, Neuberger, J, Williams, R. Evidence for expression in human liver of halothane-induced neoantigens recognized by antibodies in sera from patients with halothane hepatitis. Hepatology 1988;8:1635–41.Google Scholar
Satoh, H, Martin, BM, Schulick, AH, et al. Human anti-endoplasmic reticulum antibodies in sera of patients with halothane-induced hepatitis are directed against a trifluoroacetylated carboxylesterase. Proc Natl Acad Sci (USA) 1989;86:322–6.Google Scholar
Cote, G, Bouchard, S. Hepatotoxicity after desflurane anesthesia in a 15-month-old child with Mobius syndrome after previous exposure to isoflurane. Anesthesiology 2007;107:843–5.Google Scholar
Her, C. Acetaminophen-induced, not desflurane-induced, hepatotoxicity. Anesthesiology 2008;109:570–1.Google Scholar
Hausmann, R, Schmidt, B, Schellmann, B, et al. Differential diagnosis of postoperative liver failure in a 12-year-old child. Int J Legal Med 1996;109:210–12.Google Scholar
Jang, Y, Kim, I. Severe hepatotoxicity after sevoflurane anesthesia in a child with mild renal dysfunction. Paediatr Anaesth 2005;15:1140–4.Google Scholar
Reich, A, Everding, AS, Bulla, M, et al. Hepatitis after sevoflurane exposure in an infant suffering from primary hyperoxaluria type 1. Anesth Analg 2004;99:370–2.Google Scholar
Abdualmjid, RJ, Sergi, C. Hepatotoxic botanicals – an evidence-based systematic review. J Pharm Pharm Sci 2013;16:376404.Google Scholar
Navarro, VJ, Khan, I, Bjornsson, E, et al. Liver injury from herbal and dietary supplements. Hepatology 2017;65:363–73.Google Scholar
Rode, D. Comfrey toxicity revisited. Trends Pharmacol Sci 2002;23:497–9.Google Scholar
Zuckerman, M, Steenkamp, V, Stewart, MJ. Hepatic veno-occlusive disease as a result of a traditional remedy: confirmation of toxic pyrrolizidine alkaloids as the cause, using an in vitro technique. J Clin Pathol 2002;55:676–9.Google Scholar
Weston, CFM, Cooper, BT, Davies, JD, et al. Veno-occlusive disease of the liver secondary to ingestion of comfrey. Br Med J 1987;295:183.Google Scholar
Roulet, M, Laurini, R, Rivier, L, et al. Hepatic veno-occlusive disease in newborn infant of a woman drinking herbal tea. J Pediatr 1988;112:433–6.Google Scholar
Sperl, W, Stuppner, H, Gassner, I, et al. Reversible hepatic veno-occlusive disease in an infant after consumption of pyrrolizidine-containing herbal tea. Eur J Pediatr 1995;154:112–16.Google Scholar
Clouatre, DL. Kava kava: examining new reports of toxicity. Toxicol Lett 2004;150:8596.Google Scholar
Campo, JV, McNabb, J, Perel, JM, et al. Kava-induced fulminant hepatic failure. J Am Acad Child Adolesc Psychiatry 2002;41:631–2.Google Scholar
Anke, J, Ramzan, I. Pharmacokinetic and pharmacodynamic drug interactions with Kava (Piper methysticum Forst. f.). J Ethnopharmacol 2004;93:153–60.Google Scholar
Batchelor, WB, Heathcote, J, Wanless, IR. Chaparral-induced hepatic injury. Am J Gastroenterol 1995;90:831–3.Google Scholar
Sheikh, NM, Philen, RM, Love, LA. Chaparral-associated hepatotoxicity. Arch Intern Med 1997;157:913–19.Google Scholar
Horowitz, RS, Feldhaus, K, Dart, RC, et al. The clinical spectrum of Jin Bu Huan toxicity. Arch Intern Med 1996;156:899903.Google Scholar
Skoulidis, F, Alexander, GJ, Davies, SE. Ma huang associated acute liver failure requiring liver transplantation. Eur J Gastroenterol Hepatol 2005;17:581–4.Google Scholar
Patel, SS, Beer, S, Kearney, DL, et al. Green tea extract: a potential cause of acute liver failure. World J Gastroenterol 2013;19:5174–7.Google Scholar
Laliberte, L, Villeneuve, JP. Hepatitis after the use of germander, a herbal remedy. CMAJ 1996;154:1689–92.Google Scholar
Lekehal, M, Pessayre, D, Lereau, JM, et al. Hepatotoxicity of the herbal medicine germander: metabolic activation of its furano diterpenoids by cytochrome P450 3 A Depletes cytoskeleton-associated protein thiols and forms plasma membrane blebs in rat hepatocytes. Hepatology 1996;24:212–18.Google Scholar
Fau, D, Lekehal, M, Farrell, G, et al. Diterpenoids from germander, an herbal medicine, induce apoptosis in isolated rat hepatocytes. Gastroenterology 1997;113:1334–46.Google Scholar
Lawrenson, JA, Walls, T, Day, AS. Echinacea-induced acute liver failure in a child. J Paediatr Child Health 2014;50:841.Google Scholar
Webb, N, Hardikar, W, Cranswick, NE, et al. Probable herbal medication induced fulminant hepatic failure. J Paediatr Child Health 2005;41:530–1.Google Scholar
Zhu, Y, Li, YG, Wang, JB, et al. Causes, features, and outcomes of drug-induced liver injury in 69 children from China. Gut Liver 2015;9:525–33.Google Scholar
Goldfeld, DA, Verna, EC, Lefkowitch, J, et al. Infliximab-induced autoimmune hepatitis with successful switch to adalimumab in a patient with Crohn’s disease: the index case. Dig Dis Sci 2011;56:3386–8.Google Scholar
Ghabril, M, Bonkovsky, HL, Kum, C, et al. Liver injury from tumor necrosis factor-alpha antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol 2013;11:558–64 e553.Google Scholar
Bjornsson, ES, Gunnarsson, BI, Grondal, G, et al. Risk of drug-induced liver injury from tumor necrosis factor antagonists. Clin Gastroenterol Hepatol 2015;13:602–8.Google Scholar
French, JB, Bonacini, M, Ghabril, M, et al. Hepatotoxicity associated with the use of anti-TNF-alpha agents. Drug Saf 2016;39:199208.Google Scholar
Mostamand, S, Schroeder, S, Schenkein, J, et al. Infliximab-associated immunomediated hepatitis in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2016;63:94–7.Google Scholar
Ricciuto, A, Kamath, BM, Walters, TD, et al. New onset autoimmune hepatitis during anti-tumor necrosis factor-alpha treatment in children. J Pediatr 2018;194:128–35 e121.Google Scholar
Fathalla, BM, Goldsmith, DP, Pascasio, JM, et al. Development of autoimmune hepatitis in a child with systemic-onset juvenile idiopathic arthritis during therapy with etanercept. J Clin Rheumatol 2008;14:297–8.Google Scholar
Zimmerman, HJ. Update of hepatotoxicity due to classes of drugs in common clinical use: non-steroidal drugs, anti-inflammatory drugs, antibiotics, antihypertensives, and cardiac and psychotropic drugs. Sem Liver Dis 1990;10:322–38.Google Scholar
Donald, PR. Antituberculosis drug-induced hepatotoxicity in children. Pediatr Rep 2011;3:e16.Google Scholar
Beaudry, P, Brickman, H, Wise, M, et al. Liver enzyme disturbances during isoniazid chemoprophylaxis in children. Am Rev Resp Dis 1974;110:581–4.Google Scholar
Spyridis, P, Sinantios, C, Papadea, I, et al. Isoniazid liver injury during chemoprophylaxis in children. Arch Dis Child 1979;54:65–7.Google Scholar
Palusci, VJ, O’Hare, D, Lawrence, RM. Hepatotoxicity and transaminase measurement during isoniazid chemoprophylaxis in children. Pediatr Infect Dis J 1995;14:144–8.Google Scholar
Wu, SS, Chao, CS, Vargas, JH, et al. Isoniazid-related hepatic failure in children: a survey of liver transplantation centers. Transplantation 2007;84:173–9.Google Scholar
Frydenberg, AR, Graham, SM. Toxicity of first-line drugs for treatment of tuberculosis in children: review. Trop Med Int Health 2009;14:1329–37.Google Scholar
Chang, SH, Nahid, P, Eitzman, SR. Hepatotoxicity in children receiving isoniazid therapy for latent tuberculosis infection. J Pediatric Infect Dis Soc 2014;3:221–7.Google Scholar
Campos-Franco, J, Gonzalez-Quintela, A, Alende-Sixto, MR. Isoniazid-induced hyperacute liver failure in a young patient receiving carbamazepine. Eur J Intern Med 2004;15:396–7.Google Scholar
Indumathi, CK, Sethuraman, A, Jain, S, et al. Revised antituberculosis drug doses and hepatotoxicity in HIV negative children. Indian J Pediatr 2019;86:229–32.Google Scholar
Metushi, IG, Cai, P, Zhu, X, et al. A fresh look at the mechanism of isoniazid-induced hepatotoxicity. Clin Pharmacol Ther 2011;89:911–14.Google Scholar
Wang, P, Pradhan, K, Zhong, XB, et al. Isoniazid metabolism and hepatotoxicity. Acta Pharm Sin B 2016;6:384–92.Google Scholar
Vuilleumier, N, Rossier, MF, Chiappe, A, et al. CYP2E1 genotype and isoniazid-induced hepatotoxicity in patients treated for latent tuberculosis. Eur J Clin Pharmacol 2006;62:423–9.Google Scholar
Kyriakidis, I, Tragiannidis, A, Munchen, S, et al. Clinical hepatotoxicity associated with antifungal agents. Expert Opin Drug Saf 2017;16:149–65.Google Scholar
Lewis, JH, Zimmerman, HJ, Benson, GD, et al. Hepatic injury associated with ketoconazole therapy. Analysis of 33 cases. Gastroenterology 1984;86:503–13.Google Scholar
Egunsola, O, Adefurin, A, Fakis, A, et al. Safety of fluconazole in paediatrics: a systematic review. Eur J Clin Pharmacol 2013;69:1211–21.Google Scholar
Aghai, ZH, Mudduluru, M, Nakhla, TA, et al. Fluconazole prophylaxis in extremely low birth weight infants: association with cholestasis. J Perinatol 2006;26:550–5.Google Scholar
Benjamin, DK Jr., Hudak, ML, Duara, S, et al. Effect of fluconazole prophylaxis on candidiasis and mortality in premature infants: a randomized clinical trial. JAMA 2014;311:1742–9.Google Scholar
Schlienger, RG, Knowles, SR, Shear, NH. Lamotrigine-associated anticonvulsant hypersensitivity syndrome. Neurology 1998;51:1172–5.Google Scholar
Fayad, M, Choueiri, R, Mikati, M. Potential hepatotoxicity of lamotrigine. Pediatr Neurol 2000;22:4952.Google Scholar
Overstreet, K, Costanza, C, Behling, C, et al. Fatal progressive hepatic necrosis associated with lamotrigine treatment: a case report and literature review. Dig Dis Sci 2002;47:1921–5.Google Scholar
Arnon, R, DeVivo, D, Defelice, AR, et al. Acute hepatic failure in a child treated with lamotrigine. Pediatr Neurol 1998;18:251–2.Google Scholar
Bhayana, H, Appasani, S, Thapa, BR, et al. Lamotrigine-induced vanishing bile duct syndrome in a child. J Pediatr Gastroenterol Nutr 2012;55:e147–8.Google Scholar
Zidd, AG, Hack, JB. Pediatric ingestion of lamotrigine. Pediatr Neurol 2004;31:71–2.Google Scholar
Kremer, JM, Lee, RG, Tolman, KG. Liver histology in rheumatoid arthritis patients receiving long-term methotrexate therapy. A prospective study with baseline and sequential biopsy samples. Arthritis & Rheumatism 1989;32:121–7.Google Scholar
Kremer, JM, Furst, DE, Weinblatt, ME, et al. Significant changes in serum AST across hepatic histological grades: prospective analysis of 3 cohorts receiving methotrxate therapy for rheumatoid arthritis. J Rheumatol 1996;23:459–61.Google Scholar
Hashkes, PJ, Balistreri, WF, Bove, KE, et al. The relationship of hepatotoxic risk factors and liver histology in methotrexate therapy for juvenile rheumatoid arthritis. J Pediatr 1999;134:4752.Google Scholar
Graham, LD, Myones, BL, Rivas-Chacon, RF, et al. Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr 1992;120:468–73.Google Scholar
Kugathasam, S, Newman, AJ, Dahms, BB, et al. Liver biopsy findings in patients with juvenile rheumatoid arthritis receiving long-term, weekly methotrexate therapy. J Pediatr 1996;128:149–51.Google Scholar
Keim, D, Ragsdale, C, Heidelberger, K, et al. Hepatic fibrosis with the use of methotrexate for juvenile rheumatoid arthritis. J Rheumatol 1990;17:846–8.Google Scholar
Valentino, PL, Church, PC, Shah, PS, et al. Hepatotoxicity caused by methotrexate therapy in children with inflammatory bowel disease: a systematic review and meta-analysis. Inflamm Bowel Dis 2014;20:4759.Google Scholar
Locasciulli, A, Mura, R, Fraschini, D, et al. High-dose methotrexate administration and acute liver damage in children treated for acute lymphoblastic leukemia. A prospective study. Haematologica 1992;77:4953.Google Scholar
Fisher, MC, Cronstein, BN. Metaanalysis of methylenetetrahydrofolate reductase (MTHFR) polymorphisms affecting methotrexate toxicity. J Rheumatol 2009;36:539–45.Google Scholar
Malcolm, A, Heap, TR, Eckstein, RP, et al. Minocycline-induced liver injury. Am J Gastroenterol 1996;91:1641–3.Google Scholar
Gough, A, Chapman, S, Wagstaff, K, et al. Minocycline induced autoimmune hepatitis and systemic lupus erythematosus-like syndrome. BMJ 1996;312:169–72.Google Scholar
Bhat, G, Jordan, J Jr., Sokalski, S, et al. Minocycline-induced hepatitis with autoimmune features and neutropenia. J Clin Gastroenterol 1998;27:74–5.Google Scholar
Vo, HD, Jimenez-Rivera, C, Critch, J, et al. Distinctive features of minocycline-induced autoimmune hepatitis in children. Gastroenterology 2015;148:S996.Google Scholar
Urban, TJ, Nicoletti, P, Chalasani, N, et al. Minocycline hepatotoxicity: clinical characterization and identification of HLA-B*35:02 as a risk factor. J Hepatol 2017;67:137–44.Google Scholar
Davies, MG, Kersey, PJW. Acute hepatitis and exfoliative dermatitis associated with minocycline. BMJ 1989;298:1523–4.Google Scholar
Boudreaux, JP, Hayes, DH, Mizrahi, S, et al. Fulminant hepatic failure, hepatorenal syndrome, and necrotizing pancreatitis after minocycline hepatotoxicity. Transplant Proc 1993;25:1873.Google Scholar
Iveli, P, Noguera-Julian, A, Soler-Palacin, P, et al. [Hepatotoxicity in healthy infants exposed to nevirapine during pregnancy. Enferm Infecc Microbiol Clin 2016;34:3944.Google Scholar
McKoy, JM, Bennett, CL, Scheetz, MH, et al. Hepatotoxicity associated with long- versus short-course HIV-prophylactic nevirapine use: a systematic review and meta-analysis from the Research on Adverse Drug events And Reports (RADAR) project. Drug Saf 2009;32:147–58.Google Scholar
Ciccacci, C, Borgiani, P, Ceffa, S, et al. Nevirapine-induced hepatotoxicity and pharmacogenetics: a retrospective study in a population from Mozambique. Pharmacogenomics 2010;11:2331.Google Scholar
Ciccacci, C, Di Fusco, D, Marazzi, MC, et al. Association between CYP2B6 polymorphisms and nevirapine-induced SJS/TEN: a pharmacogenetics study. Eur J Clin Pharmacol 2013;69:1909–16.Google Scholar
Singh, H, Lata, S, Dhole, TN, et al. Occurrence of CYP2B6 516G>T polymorphism in patients with ARV-associated hepatotoxicity. Mol Genet Genomic Med 2019;7:e00598.Google Scholar
Agundez, JA, Lucena, MI, Martinez, C, et al. Assessment of nonsteroidal anti-inflammatory drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2011;7:817–28.Google Scholar
Titchen, T, Cranswick, N, Beggs, S. Adverse drug reactions to nonsteroidal anti-inflammatory drugs, COX-2 inhibitors and paracetamol in a paediatric hospital. Br J Clin Pharmacol 2005;59:718–23.Google Scholar
Whittaker, SJ, Amar, JN, Wanless, IR, et al. Sulindac hepatotoxicity. Gut 1982;23:875–7.Google Scholar
Boelsterli, UA. Diclofenac-induced liver injury: a paradigm of idiosyncratic drug toxicity. Toxicol Appl Pharmacol 2003;192:307–22.Google Scholar
Merlani, G, Fox, M, Oehen, HP, et al. Fatal hepatoxicity secondary to nimesulide. Eur J Clin Pharmacol 2001;57:321–6.Google Scholar
Bessone, F, Colombato, L, Fassio, E, et al. The spectrum of nimesulide-induced-hepatotoxicity. An overview. Antiinflamm Antiallergy Agents Med Chem 2010;9:355–65.Google Scholar
Marotta, PJ, Roberts, EA. Pemoline hepatotoxicity in children. J Pediatr 1998;132:894–7.Google Scholar
Rosh, JR, Dellert, SF, Narkewicz, M, et al. Four cases of severe hepatotoxicity associated with pemoline: possible autoimmune pathogenesis. Pediatrics 1998;101:921–3.Google Scholar
Gresser, U. Amoxicillin-clavulanic acid therapy may be associated with severe side effects – review of the literature. Eur J Med Res 2001;6:139–49.Google Scholar
deLemos, AS, Ghabril, M, Rockey, DC, et al. Amoxicillin-clavulanate-induced liver injury. Dig Dis Sci 2016;61:2406–16.Google Scholar
Stricker, BH, Van den Broek, JW, Keuning, J, et al. Cholestatic hepatitis due to antibacterial combination of amoxicillin and clavulanic acid (augmentin). Dig Dis Sci 1989;34:1576–80.Google Scholar
Chawla, A, Kahn, E, Yunis, EJ, et al. Rapidly progressive cholstasis: an unusual reaction to amoxicillin/clavulinic acid in a child. J Pediatr 2000;136:121–3.Google Scholar
Yu, MK, Yu, MC, Lee, F. Association of DRESS syndrome with chylous ascites. Nephrol Dial Transplant 2006;21:3301–3.Google Scholar
Kumar, A, Sood, V, Khanna, R, et al. Clinical spectrum and outcome of pediatric drug induced liver injury. Indian J Pediatr 2018;85:676–8.Google Scholar
Fontana, RJ, Shakil, AO, Greenson, JK, et al. Acute liver failure due to amoxicillin and amoxicillin/clavulanate. Dig Dis Sci 2005;50:1785–90.Google Scholar
Daly, AK, Donaldson, PT, Bhatnagar, P, et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat Genet 2009;41:816–19.Google Scholar
Lucena, MI, Molokhia, M, Shen, Y, et al. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011;141:338–47.Google Scholar
Roberts, EA, Spielberg, SP, Goldbach, M, et al. Phenobarbital hepatotoxicity in an 8-month-old infant. J Hepatol 1990;10:235–9.Google Scholar
Pinna, AP, Locci, G, Furno, M, et al. DILI (drug induced liver injury) in a 9-month-old infant: a rare case of phenobarbital-induced hepatotoxicity. J Pediatr Neonat Individ Med 2013;2:93–5.Google Scholar
Li, AM, Nelson, EA, Hon, EK, et al. Hepatic failure in a child with anti-epileptic hypersensitivity syndrome. J Paediatr Child Health 2005;41:218–20.Google Scholar
Bessmertny, O, Hatton, RC, Gonzalez-Peralta, RP. Antiepileptic hypersensitivity syndrome in children. Ann Pharmacother 2001;35:533–8.Google Scholar
Mullick, FG, Ishak, KG. Hepatic injury associated with diphenylhydantoin therapy. Am J Clin Pathol 1980;74:442–52.Google Scholar
Spielberg, SP, Gordon, GB, Blake, DA, et al. Anticonvulsant toxicity in vitro: possible role of arene oxides. J Pharmacol Exp Ther 1981;217:386–9.Google Scholar
Spielberg, SP, Gordon, GB, Blake, DA, et al. Predisposition to phenytoin hepatotoxicity assessed in vitro. N Engl J Med 1981;305:722–7.Google Scholar
Akmal, A, Kung, J. Propylthiouracil, methimazole, and carbimazole-related hepatotoxicity. Expert Opin Drug Saf 2014;13:13971406.Google Scholar
Rivkees, SA, Mattison, DR. Propylthiouracil (PTU) hepatotoxicity in children and recommendations for discontinuation of use. Int J Pediatr Endocrinol 2009;2009:132041.Google Scholar
Rivkees, SA, Szarfman, A. Dissimilar hepatotoxicity profiles of propylthiouracil and methimazole in children. J Clin Endocrinol Metab 2010;95:3260–7.Google Scholar
Maggiore, G, Larizza, D, Lorini, R, et al. PTU hepatotoxicity mimicking autoimmune chronic active hepatitis in a girl. J Pediatr Gastroenterol Nutr 1989;8:547–8.Google Scholar
Hayashida, CY, Duarte, AJ, Sato, AE, et al. Neonatal hepatitis and lymphocyte sensitization by placental transfer of propylthiouracil. J Endocrinol Invest 1990;13:937–41.Google Scholar
Loomba-Albrecht, LA, Bremer, AA, Wong, A, et al. Neonatal cholestasis due to hyperthyroidism. J Pediatr Gastroenterol Nutr 2012;54:433–4.Google Scholar
Zane, LT, Leyden, WA, Marqueling, AL, et al. A population-based analysis of laboratory abnormalities during isotretinoin therapy for acne vulgaris. Arch Dermatol 2006;142:1016–22.Google Scholar
Guzman Rojas, P, Gallegos Lopez, R, Ciliotta Chehade, A, et al. Autoimmune hepatitis induced by isotretionine. Rev Gastroenterol Peru 2016;36:86–9.Google Scholar
Fallon, MB, Boyer, JL. Hepatic toxicity of vitamin A and synthetic retinoids. J Gastroenterol Hepatol 1990;5:334–42.Google Scholar
Kumra, S, Herion, D, Jacobsen, LK, et al. Case study: risperidone-induced hepatotoxicity in pediatric patients. J Am Acad Child Adolesc Psychiatry 1997;36:701–5.Google Scholar
Krebs, S, Dormann, H, Muth-Selbach, U, et al. Risperidone-induced cholestatic hepatitis. Eur J Gastroenterol Hepatol 2001;13:67–9.Google Scholar
Copur, M, Erdogan, A. Risperidone rechallenge for marked liver function test abnormalities in an autistic child. Recent Pat Endocr Metab Immune Drug Discov 2011;5:237–9.Google Scholar
Bjornsson, ES. Hepatotoxicity of statins and other lipid-lowering agents. Liver Int 2017;37:173–8.Google Scholar
Desai, NK, Mendelson, MM, Baker, A, et al. Hepatotoxicity of statins as determined by serum alanine aminotransferase in a pediatric cohort with dyslipidemia. J Pediatr Gastroenterol Nutr 2019;68:175–81.Google Scholar
Olah, AV, Szabo, GP, Varga, J, et al. Relation between biomarkers and clinical severity in patients with Smith-Lemli-Opitz syndrome. Eur J Pediatr 2013;172:623–30.Google Scholar
Shear, NH, Spielberg, SP, Grant, DM, et al. Differences in metabolism of sulfonamides predisposing to idiosyncratic toxicity. Ann Intern Med 1986;105:179–84.Google Scholar
Besnard, M, Debray, D, Durand, P, et al. Fulminant hepatitis in two children treated with sulfasalazine for Crohn disease. Arch Pediatr 1999;6:643–6.Google Scholar
Karpman, E, Kurzrock, EA. Adverse reactions of nitrofurantoin, trimethoprim and sulfamethoxazole in children. J Urol 2004;172:448–53.Google Scholar
Bucaretchi, F, Vicente, DC, Pereira, RM, et al. Dapsone hypersensitivity syndrome in an adolescent during treatment of leprosy. Rev Inst Med Trop Sao Paulo 2004;46:331–4.Google Scholar
Rieder, MJ, Uetrecht, J, Shear, NH, et al. Diagnosis of sulfonamide hypersensitivity reactions by in-vitro “rechallenge” with hydroxylamine metabolites. Ann Intern Med 1989;110:286–9.Google Scholar
Cribb, AE, Spielberg, SP. Hepatic microsomal metabolism of sulfamethoxazole to the hydroxylamine. Drug Metab Dispos 1990;18:784–7.Google Scholar
Sanderson, JP, Naisbitt, DJ, Park, BK. Role of bioactivation in drug-induced hypersensitivity reactions. AAPS J 2006;8:E5564.Google Scholar
Ogese, MO, Faulkner, L, Jenkins, RE, et al. Characterization of drug-specific signaling between primary human hepatocytes and immune cells. Toxicol Sci 2017;158:7689.Google Scholar
Ogese, MO, Jenkins, RE, Adair, K, et al. Exosomal transport of hepatocyte-derived drug-modified proteins to the immune system. Hepatology 2019;70(5):1732–49.Google Scholar
Sztajnkrycer, MD. Valproic acid toxicity: overview and management. J Toxiocl Clin Toxicol 2002;40:789801.Google Scholar
Suchy, FJ, Balistreri, WF, Buchino, J, et al. Acute hepatic failure associated with the use of sodium valproate. Report of two fatal cases. N Engl J Med 1979;300:962–6.Google Scholar
Zimmerman, HJ, Ishak, KG. Valproate-induced hepatic injury: analysis of 23 fatal cases. Hepatology 1982;2:591–7.Google Scholar
Koenig, SA, Siemes, H, Blaker, F, et al. Severe hepatotoxicity during valproate therapy: an update and report of eight new fatalities. Epilepsia 1994;35:1005–15.Google Scholar
Star, K, Edwards, IR, Choonara, I. Valproic acid and fatalities in children: a review of individual case safety reports in VigiBase. PLoS One 2014;9:e108970.Google Scholar
Price, KE, Pearce, RE, Garg, UC, et al. Effects of valproic acid on organic acid metabolism in children: a metabolic profiling study. Clin Pharmacol Ther 2011;89:867–74.Google Scholar
McCall, M, Bourgeois, JA. Valproic acid-induced hyperammonemia: a case report. J Clin Psychopharmacol 2004;24:521–6.Google Scholar
Gerstner, T, Buesing, D, Longin, E, et al. Valproic acid induced encephalopathy – 19 new cases in Germany from 1994 to 2003 – A side effect associated to VPA-therapy not only in young children. Seizure 2006;15:443–8.Google Scholar
Ghodke-Puranik, Y, Thorn, CF, Lamba, JK, et al. Valproic acid pathway: pharmacokinetics and pharmacodynamics. Pharmacogenet Genomics 2013;23:236–41.Google Scholar
Li, X, Norwood, DL, Mao, L-F, et al. Mitochondrial metabolism of valproic acid. Biochemistry 1991;30:388–94.Google Scholar
Kiang, TK, Ho, PC, Anari, MR, et al. Contribution of CYP2C9, CYP2A6, and CYP2B6 to valproic acid metabolism in hepatic microsomes from individuals with the CYP2C9*1/*1 genotype. Toxicol Sci 2006;94:261–71.Google Scholar
Gopaul, S, Farrell, K, Abbott, F. Effects of age and polytherapy, risk factors of valproic acid (VPA) hepatotoxicity, on the excretion of thiol conjugates of (E)-2,4-diene VPA in people with epilepsy taking VPA. Epilepsia 2003;44:322–8.Google Scholar
Eadie, MJ, McKinnon, GE, Dunstan, PR, et al. Valproate metabolism during hepatotoxicity associated with the drug. Quart J Med 1990;77:1229–40.Google Scholar
Kossak, BD, Schmidt-Sommerfeld, E, Schoeller, DA, et al. Impaired fatty acid oxidation in children on valproic acid and the effect of L-carnitine. Neurology 1993;43:2362–8.Google Scholar
Tong, V, Teng, XW, Chang, TK, et al. Valproic acid I: time course of lipid peroxidation biomarkers, liver toxicity, and valproic acid metabolite levels in rats. Toxicol Sci 2005;86:427–35.Google Scholar
Tong, V, Teng, XW, Chang, TK, et al. Valproic acid II: effects on oxidative stress, mitochondrial membrane potential, and cytotoxicity in glutathione-depleted rat hepatocytes. Toxicol Sci 2005;86:436–43.Google Scholar
McCarver, DG, Hines, RN. The ontogeny of human drug-metabolizing enzymes: phase II conjugation enzymes and regulatory mechanisms. J Pharmacol Exp Ther 2002;300:361–6.Google Scholar
Xing, Y, Yang, L, Wang, L, et al. Systematic screening for polymorphisms within the UGT1A6 gene in three Chinese populations and function prediction through structural modeling. Pharmacogenomics 2009;10:741–52.Google Scholar
Konig, SA, Schenk, M, Sick, C, et al. Fatal liver failure associated with valproate therapy in a patient with Friedreich’s disease: review of valproate hepatotoxicity in adults. Epilepsia 1999;40:1036–40.Google Scholar
Schwabe, MJ, Dobyns, WB, Burke, B, et al. Valproate-induced liver failure in one of two siblings with Alpers disease. Pediatr Neurol 1997;16:337–43.Google Scholar
Kayihan, N, Nennesmo, I, Ericzon, BG, et al. Fatal deterioration of neurological disease after orthotopic liver transplantation for valproic acid-induced liver damage. Pediatr Transplant 2000;4:211–14.Google Scholar
Vantroys, E, Smet, J, Vanlander, AV, et al. Severe hepatopathy and neurological deterioration after start of valproate treatment in a 6-year-old child with mitochondrial tryptophanyl-tRNA synthetase deficiency. Orphanet J Rare Dis 2018;13:80.Google Scholar
Stewart, JD, Horvath, R, Baruffini, E, et al. Polymerase gamma gene POLG determines the risk of sodium valproate-induced liver toxicity. Hepatology 2010;52:1791–6.Google Scholar
Li, S, Guo, J, Ying, Z, et al. Valproic acid-induced hepatotoxicity in Alpers syndrome is associated with mitochondrial permeability transition pore opening-dependent apoptotic sensitivity in an induced pluripotent stem cell model. Hepatology 2015;61:1730–9.Google Scholar
Saneto, RP, Lee, IC, Koenig, MK, et al. POLG DNA testing as an emerging standard of care before instituting valproic acid therapy for pediatric seizure disorders. Seizure 2010;19:140–6.Google Scholar
Beghi, E, Bizzi, A, Codegoni, AM, et al. Valproate, carnitine metabolism, and biochemical indicators of liver function. Epilepsia 1990;31:346–52.Google Scholar
Lheureux, PE, Hantson, P. Carnitine in the treatment of valproic acid-induced toxicity. Clin Toxicol (Phila) 2009;47:101–11.Google Scholar
Bohan, TP, Helton, E, McDonald, I, et al. Effect of L-carnitine treatment for valproate-induced hepatotoxicity. Neurology 2001;56:1405–9.Google Scholar
Squires, RH, Dhawan, A, Alonso, E, et al. Intravenous N-acetylcysteine in pediatric patients with nonacetaminophen acute liver failure: a placebo-controlled clinical trial. Hepatology 2013;57:1542–9.Google Scholar
Suzuki, A, Brunt, EM, Kleiner, DE, et al. The use of liver biopsy evaluation in discrimination of idiopathic autoimmune hepatitis versus drug-induced liver injury. Hepatology 2011;54:931–9.Google Scholar
Naranjo, CA, Busto, U, Sellers, EM, et al. A method for estimating the probability of adverse drug reactions. Clin Pharmacol Ther 1981;30:239–45.Google Scholar
Garcia-Cortes, M, Stephens, C, Lucena, MI, et al. Causality assessment methods in drug induced liver injury: strengths and weaknesses. J Hepatol 2011;55:683–91.Google Scholar
Gallagher, RM, Kirkham, JJ, Mason, JR, et al. Development and inter-rater reliability of the Liverpool adverse drug reaction causality assessment tool. PLoS One 2011;6:e28096.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×