Semin Liver Dis 2014; 34(02): 123-133
DOI: 10.1055/s-0034-1375954
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Genetic Basis of Drug-Induced Liver Injury: Present and Future

Thomas J. Urban
1   Center for Human Genome Variation, Duke University Medical Center, Durham, North Carolina
,
Ann K. Daly
2   Institute of Cellular Medicine, Newcastle University Medical School, Newcastle upon Tyne, United Kingdom
,
Guruprasad P. Aithal
3   National Institute for Health Research Nottingham Digestive Diseases Biomedical research Unit, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
› Author Affiliations
Further Information

Publication History

Publication Date:
31 May 2014 (online)

Abstract

There is considerable evidence that susceptibility to idiosyncratic drug-induced liver injury (DILI) is genetically determined. Though genetic associations with DILI have been reported since the 1980s, the development of genome-wide association studies has enabled genetic risk factors for DILI, in common with other diseases, to be detected and confirmed more confidently. Human leukocyte antigen (HLA) genotype has been demonstrated to be a strong risk factor for development of DILI with a range of drugs and the underlying mechanism, probably involving presentation of a drug-peptide complex to T cells is increasingly well understood. However, specific HLA alleles are not associated with all forms of DILI and non-HLA genetic risk factors, especially those relating to drug disposition, also appear to contribute. For some drugs, there is evidence of a dual role for HLA and drug metabolism genes. Though the associations with non-HLA genes have been less well replicated than the HLA associations, there is increasing evidence that drug metabolism genes such as NAT2 and UGT2B7 contribute to some forms of DILI. Translating current genetic findings on DILI susceptibility to the clinic has been relatively slow, but some progress is now being made. In the future, DNA sequencing may lead to the identification of rare variants that contribute to DILI. Developments in the related area of epigenomics and in the development of improved models for DILI by use of genetically defined induced pluripotent stem cells should improve understanding of the biology of DILI and inform drug development.

 
  • References

  • 1 Wilke RA, Lin DW, Roden DM , et al. Identifying genetic risk factors for serious adverse drug reactions: current progress and challenges. Nat Rev Drug Discov 2007; 6 (11) 904-916
  • 2 Ostapowicz G, Fontana RJ, Schiødt FV , et al; U.S. Acute Liver Failure Study Group. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann Intern Med 2002; 137 (12) 947-954
  • 3 Meyer UA. Pharmacogenetics - five decades of therapeutic lessons from genetic diversity. Nat Rev Genet 2004; 5 (9) 669-676
  • 4 Urban TJ, Shen Y, Stolz A , et al; Drug-Induced Liver Injury Network; DILIGEN; EUDRAGENE; Spanish DILI Registry; International Serious Adverse Events Consortium. Limited contribution of common genetic variants to risk for liver injury due to a variety of drugs. Pharmacogenet Genomics 2012; 22 (11) 784-795
  • 5 Wellcome Trust Case Control C ; Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007; 447 (7145) 661-678
  • 6 McCarthy MI, Abecasis GR, Cardon LR , et al. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet 2008; 9 (5) 356-369
  • 7 Daly AK, Donaldson PT, Bhatnagar P , et al; 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 (7) 816-819
  • 8 Monshi MM, Faulkner L, Gibson A , et al. Human leukocyte antigen (HLA)-B*57:01-restricted activation of drug-specific T cells provides the immunological basis for flucloxacillin-induced liver injury. Hepatology 2013; 57 (2) 727-739
  • 9 Wuillemin N, Adam J, Fontana S, Krähenbühl S, Pichler WJ, Yerly D. HLA haplotype determines hapten or p-i T cell reactivity to flucloxacillin. J Immunol 2013; 190 (10) 4956-4964
  • 10 Hautekeete ML, Horsmans Y, Van Waeyenberge C , et al. HLA association of amoxicillin-clavulanate—induced hepatitis. Gastroenterology 1999; 117 (5) 1181-1186
  • 11 O'Donohue J, Oien KA, Donaldson P , et al. Co-amoxiclav jaundice: clinical and histological features and HLA class II association. Gut 2000; 47 (5) 717-720
  • 12 Lucena MI, Molokhia M, Shen Y , et al; Spanish DILI Registry; EUDRAGENE; DILIN; DILIGEN; International SAEC. Susceptibility to amoxicillin-clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 2011; 141 (1) 338-347
  • 13 Donaldson PT, Daly AK, Henderson J , et al. Human leucocyte antigen class II genotype in susceptibility and resistance to co-amoxiclav-induced liver injury. J Hepatol 2010; 53 (6) 1049-1053
  • 14 Kindmark A, Jawaid A, Harbron CG , et al. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J 2008; 8 (3) 186-195
  • 15 Morgan MY, Reshef R, Shah RR, Oates NS, Smith RL, Sherlock S. Impaired oxidation of debrisoquine in patients with perhexiline liver injury. Gut 1984; 25 (10) 1057-1064
  • 16 Fromenty B, Pessayre D. Inhibition of mitochondrial beta-oxidation as a mechanism of hepatotoxicity. Pharmacol Ther 1995; 67 (1) 101-154
  • 17 Shah RR. Can pharmacogenetics help rescue drugs withdrawn from the market?. Pharmacogenomics 2006; 7 (6) 889-908
  • 18 Pachkoria K, Lucena MI, Ruiz-Cabello F, Crespo E, Cabello MR, Andrade RJ ; Spanish Group for the Study of Drug-Induced Liver Disease (Grupo de Estudio para las Hepatopatías Asociadas a Medicamentos). Genetic polymorphisms of CYP2C9 and CYP2C19 are not related to drug-induced idiosyncratic liver injury (DILI). Br J Pharmacol 2007; 150 (6) 808-815
  • 19 Aithal GP, Day CP, Leathart JBS, Daly AK. Relationship of polymorphism in CYP2C9 to genetic susceptibility to diclofenac-induced hepatitis. Pharmacogenetics 2000; 10 (6) 511-518
  • 20 Markova SM, De Marco T, Bendjilali N , et al. Association of CYP2C9*2 with bosentan-induced liver injury. Clin Pharmacol Ther 2013; 94 (6) 678-686
  • 21 Ariyoshi N, Iga Y, Hirata K , et al. Enhanced susceptibility of HLA-mediated ticlopidine-induced idiosyncratic hepatotoxicity by CYP2B6 polymorphism in Japanese. Drug Metab Pharmacokinet 2010; 25 (3) 298-306
  • 22 Yimer G, Amogne W, Habtewold A , et al. High plasma efavirenz level and CYP2B6*6 are associated with efavirenz-based HAART-induced liver injury in the treatment of naïve HIV patients from Ethiopia: a prospective cohort study. Pharmacogenomics J 2012; 12 (6) 499-506
  • 23 Metushi IG, Sanders C, Lee WM, Uetrecht J ; Acute Liver Study Group. Detection of anti-isoniazid and anti-cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Hepatology 2014; 59 (3) 1084-1093
  • 24 Huang YS, Chern HD, Su WJ , et al. Cytochrome P450 2E1 genotype and the susceptibility to antituberculosis drug-induced hepatitis. Hepatology 2003; 37 (4) 924-930
  • 25 Cho HJ, Koh WJ, Ryu YJ , et al. Genetic polymorphisms of NAT2 and CYP2E1 associated with antituberculosis drug-induced hepatotoxicity in Korean patients with pulmonary tuberculosis. Tuberculosis (Edinb) 2007; 87 (6) 551-556
  • 26 Kim SH, Kim SH, Bahn JW , et al. Genetic polymorphisms of drug-metabolizing enzymes and anti-TB drug-induced hepatitis. Pharmacogenomics 2009; 10 (11) 1767-1779
  • 27 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 (6) 423-429
  • 28 Yamada S, Tang M, Richardson K , et al. Genetic variations of NAT2 and CYP2E1 and isoniazid hepatotoxicity in a diverse population. Pharmacogenomics 2009; 10 (9) 1433-1445
  • 29 Bose PD, Sarma MP, Medhi S, Das BC, Husain SA, Kar P. Role of polymorphic N-acetyl transferase2 and cytochrome P4502E1 gene in antituberculosis treatment-induced hepatitis. J Gastroenterol Hepatol 2011; 26 (2) 312-318
  • 30 Santos NP, Callegari-Jacques SM, Ribeiro Dos Santos AK , et al. N-acetyl transferase 2 and cytochrome P450 2E1 genes and isoniazid-induced hepatotoxicity in Brazilian patients. Int J Tuberc Lung Dis 2013; 17 (4) 499-504
  • 31 Gupta VH, Amarapurkar DN, Singh M , et al. Association of N-acetyltransferase 2 and cytochrome P450 2E1 gene polymorphisms with antituberculosis drug-induced hepatotoxicity in Western India. J Gastroenterol Hepatol 2013; 28 (8) 1368-1374
  • 32 Regan SL, Maggs JL, Hammond TG, Lambert C, Williams DP, Park BK. Acyl glucuronides: the good, the bad and the ugly. Biopharm Drug Dispos 2010; 31 (7) 367-395
  • 33 Acuña G, Foernzler D, Leong D , et al. Pharmacogenetic analysis of adverse drug effect reveals genetic variant for susceptibility to liver toxicity. Pharmacogenomics J 2002; 2 (5) 327-334
  • 34 Daly AK, Aithal GP, Leathart JB, Swainsbury RA, Dang TS, Day CP. Genetic susceptibility to diclofenac-induced hepatotoxicity: contribution of UGT2B7, CYP2C8, and ABCC2 genotypes. Gastroenterology 2007; 132 (1) 272-281
  • 35 Metushi IG, Cai P, Zhu X, Nakagawa T, Uetrecht JP. A fresh look at the mechanism of isoniazid-induced hepatotoxicity. Clin Pharmacol Ther 2011; 89 (6) 911-914
  • 36 Eichelbaum M, Musch E, Castroparra M, Vonsassen W. Isoniazid hepatotoxicity in relation to acetylator phenotype and isoniazid metabolism. Br J Clin Pharmacol 1982; 14 (4) 575 –P576
  • 37 Grönhagen-Riska C, Hellstrom PE, Fröseth B. Predisposing factors in hepatitis induced by isoniazid-rifampin treatment of tuberculosis. Am Rev Respir Dis 1978; 118 (3) 461-466
  • 38 Dickinson DS, Bailey WC, Hirschowitz BI, Soong SJ, Eidus L, Hodgkin MM. Risk factors for isoniazid (NIH)-induced liver dysfunction. J Clin Gastroenterol 1981; 3 (3) 271-279
  • 39 Mitchell JR, Thorgeirsson UP, Black M , et al. Increased incidence of isoniazid hepatitis in rapid acetylators: possible relation to hydranize metabolites. Clin Pharmacol Ther 1975; 18 (1) 70-79
  • 40 Ohno M, Yamaguchi I, Yamamoto I , et al. Slow N-acetyltransferase 2 genotype affects the incidence of isoniazid and rifampicin-induced hepatotoxicity. Int J Tuberc Lung Dis 2000; 4 (3) 256-261
  • 41 Huang YS, Chern HD, Su WJ , et al. Polymorphism of the N-acetyltransferase 2 gene as a susceptibility risk factor for antituberculosis drug-induced hepatitis. Hepatology 2002; 35 (4) 883-889
  • 42 Lee SW, Chung LS, Huang HH, Chuang TY, Liou YH, Wu LS. NAT2 and CYP2E1 polymorphisms and susceptibility to first-line anti-tuberculosis drug-induced hepatitis. Int J Tuberc Lung Dis 2010; 14 (5) 622-626
  • 43 Ho HT, Wang TH, Hsiong CH , et al. The NAT2 tag SNP rs1495741 correlates with the susceptibility of antituberculosis drug-induced hepatotoxicity. Pharmacogenet Genomics 2013; 23 (4) 200-207
  • 44 Possuelo LG, Castelan JA, de Brito TC , et al. Association of slow N-acetyltransferase 2 profile and anti-TB drug-induced hepatotoxicity in patients from Southern Brazil. Eur J Clin Pharmacol 2008; 64 (7) 673-681
  • 45 Bozok Cetintaş V, Erer OF, Kosova B , et al. Determining the relation between N-acetyltransferase-2 acetylator phenotype and antituberculosis drug induced hepatitis by molecular biologic tests. Tuberk Toraks 2008; 56 (1) 81-86
  • 46 Chamorro JG, Castagnino JP, Musella RM , et al. Sex, ethnicity, and slow acetylator profile are the major causes of hepatotoxicity induced by antituberculosis drugs. J Gastroenterol Hepatol 2013; 28 (2) 323-328
  • 47 Leiro-Fernandez V, Valverde D, Vázquez-Gallardo R , et al. N-acetyltransferase 2 polymorphisms and risk of anti-tuberculosis drug-induced hepatotoxicity in Caucasians. Int J Tuberc Lung Dis 2011; 15 (10) 1403-1408
  • 48 Saukkonen JJ, Cohn DL, Jasmer RM , et al; ATS (American Thoracic Society) Hepatotoxicity of Antituberculosis Therapy Subcommittee. An official ATS statement: hepatotoxicity of antituberculosis therapy. Am J Respir Crit Care Med 2006; 174 (8) 935-952
  • 49 Roy B, Chowdhury A, Kundu S , et al. Increased risk of antituberculosis drug-induced hepatotoxicity in individuals with glutathione S-transferase M1 'null' mutation. J Gastroenterol Hepatol 2001; 16 (9) 1033-1037
  • 50 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; 47 (1) 128-134
  • 51 Leiro V, Fernández-Villar A, Valverde D , et al. Influence of glutathione S-transferase M1 and T1 homozygous null mutations on the risk of antituberculosis drug-induced hepatotoxicity in a Caucasian population. Liver Int 2008; 28 (6) 835-839
  • 52 Simon T, Becquemont L, Mary-Krause M , et al. Combined glutathione-S-transferase M1 and T1 genetic polymorphism and tacrine hepatotoxicity. Clin Pharmacol Ther 2000; 67 (4) 432-437
  • 53 Watanabe I, Tomita A, Shimizu M , et al. A study to survey susceptible genetic factors responsible for troglitazone-associated hepatotoxicity in Japanese patients with type 2 diabetes mellitus. Clin Pharmacol Ther 2003; 73 (5) 435-455
  • 54 Lucena MI, Andrade RJ, Martínez C , et al; Spanish Group for the Study of Drug-Induced Liver Disease. Glutathione S-transferase m1 and t1 null genotypes increase susceptibility to idiosyncratic drug-induced liver injury. Hepatology 2008; 48 (2) 588-596
  • 55 Okada R, Maeda K, Nishiyama T , et al. Involvement of different human glutathione transferase isoforms in the glutathione conjugation of reactive metabolites of troglitazone. Drug Metab Dispos 2011; 39 (12) 2290-2297
  • 56 Lucena MI, García-Martín E, Andrade RJ , et al. Mitochondrial superoxide dismutase and glutathione peroxidase in idiosyncratic drug-induced liver injury. Hepatology 2010; 52 (1) 303-312
  • 57 Andrews E, Armstrong M, Tugwood J , et al. A role for the pregnane X receptor in flucloxacillin-induced liver injury. Hepatology 2010; 51 (5) 1656-1664
  • 58 Kuehl P, Zhang J, Lin Y , et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001; 27 (4) 383-391
  • 59 Li T, Yu RT, Atkins AR, Downes M, Tukey RH, Evans RM. Targeting the pregnane X receptor in liver injury. Expert Opin Ther Targets 2012; 16 (11) 1075-1083
  • 60 Li F, Lu J, Cheng J , et al. Human PXR modulates hepatotoxicity associated with rifampicin and isoniazid co-therapy. Nat Med 2013; 19 (4) 418-420
  • 61 Ahmadian M, Suh JM, Hah N , et al. PPARγ signaling and metabolism: the good, the bad and the future. Nat Med 2013; 19 (5) 557-566
  • 62 Geier A, Wagner M, Dietrich CG, Trauner M. Principles of hepatic organic anion transporter regulation during cholestasis, inflammation and liver regeneration. Biochim Biophys Acta 2007; 1773 (3) 283-308
  • 63 Noe J, Kullak-Ublick GA, Jochum W , et al. Impaired expression and function of the bile salt export pump due to three novel ABCB11 mutations in intrahepatic cholestasis. J Hepatol 2005; 43 (3) 536-543
  • 64 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 (1) 47-60
  • 65 Bhatnagar P, Day CP, Aithal G , et al. Genetic variants of hepatic transporters and susceptibility to drug induced liver injury. Toxicology 2008; 253 (1–3) 10
  • 66 Haenisch S, Zimmermann U, Dazert E , et al. Influence of polymorphisms of ABCB1 and ABCC2 on mRNA and protein expression in normal and cancerous kidney cortex. Pharmacogenomics J 2007; 7 (1) 56-65
  • 67 Choi JH, Ahn BM, Yi J , et al. MRP2 haplotypes confer differential susceptibility to toxic liver injury. Pharmacogenet Genomics 2007; 17 (6) 403-415
  • 68 Haas DW, Bartlett JA, Andersen JW , et al; Adult AIDS Clinical Trials Group. Pharmacogenetics of nevirapine-associated hepatotoxicity: an Adult AIDS Clinical Trials Group collaboration. Clin Infect Dis 2006; 43 (6) 783-786
  • 69 Ritchie MD, Haas DW, Motsinger AA , et al. Drug transporter and metabolizing enzyme gene variants and nonnucleoside reverse-transcriptase inhibitor hepatotoxicity. Clin Infect Dis 2006; 43 (6) 779-782
  • 70 Yuan J, Guo S, Hall D , et al; Nevirapine Toxicogenomics Study Team. Toxicogenomics of nevirapine-associated cutaneous and hepatic adverse events among populations of African, Asian, and European descent. AIDS 2011; 25 (10) 1271-1280
  • 71 Holmes MV, Casas JP, Hingorani AD. Putting genomics into practice. BMJ 2011; 343: d4953
  • 72 Aithal GP, Watkins PB, Andrade RJ , et al. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther 2011; 89 (6) 806-815
  • 73 Olson H, Betton G, Robinson D , et al. Concordance of the toxicity of pharmaceuticals in humans and in animals. Regul Toxicol Pharmacol 2000; 32 (1) 56-67
  • 74 Mallal S, Phillips E, Carosi G , et al; PREDICT-1 Study Team. HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med 2008; 358 (6) 568-579
  • 75 El Sherrif Y, Potts JR, Howard MR , et al. Hepatotoxicity from anabolic androgenic steroids marketed as dietary supplements: contribution from ATP8B1/ABCB11 mutations?. Liver Int 2013; 33 (8) 1266-1270
  • 76 Alfirevic A, Pirmohamed M. Predictive genetic testing for drug-induced liver injury: considerations of clinical utility. Clin Pharmacol Ther 2012; 92 (3) 376-380
  • 77 Chen P, Lin JJ, Lu CS , et al; Taiwan SJS Consortium. Carbamazepine-induced toxic effects and HLA-B*1502 screening in Taiwan. N Engl J Med 2011; 364 (12) 1126-1133
  • 78 Hung SI, Chung WH, Liou LB , et al. HLA-B*5801 allele as a genetic marker for severe cutaneous adverse reactions caused by allopurinol. Proc Natl Acad Sci U S A 2005; 102 (11) 4134-4139
  • 79 Kazeem GR, Cox C, Aponte J , et al. High-resolution HLA genotyping and severe cutaneous adverse reactions in lamotrigine-treated patients. Pharmacogenet Genomics 2009; 19 (9) 661-665
  • 80 Yip VL, Marson AG, Jorgensen AL, Pirmohamed M, Alfirevic A. HLA genotype and carbamazepine-induced cutaneous adverse drug reactions: a systematic review. Clin Pharmacol Ther 2012; 92 (6) 757-765
  • 81 Zhang FR, Liu H, Irwanto A , et al. HLA-B*13:01 and the dapsone hypersensitivity syndrome. N Engl J Med 2013; 369 (17) 1620-1628
  • 82 Dubios PCA. Thiopurine induced pancreatitis genetics working group. The risk of azathioprine-induced pancreatitis depends on genetic variants in the HLA gene region. Gut 2011; 60: A60
  • 83 Goldstein DB. Common genetic variation and human traits. N Engl J Med 2009; 360 (17) 1696-1698
  • 84 Maher B. Personal genomes: The case of the missing heritability. Nature 2008; 456 (7218) 18-21
  • 85 Chung WH, Hung SI, Hong HS , et al. Medical genetics: a marker for Stevens-Johnson syndrome. Nature 2004; 428 (6982) 486
  • 86 Hughes AR, Spreen WR, Mosteller M , et al. Pharmacogenetics of hypersensitivity to abacavir: from PGx hypothesis to confirmation to clinical utility. Pharmacogenomics J 2008; 8 (6) 365-374
  • 87 Liu R, Paxton WA, Choe S , et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 1996; 86 (3) 367-377
  • 88 Samson M, Libert F, Doranz BJ , et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 1996; 382 (6593) 722-725
  • 89 Li B, Leal SM. Methods for detecting associations with rare variants for common diseases: application to analysis of sequence data. Am J Hum Genet 2008; 83 (3) 311-321
  • 90 Bamshad MJ, Ng SB, Bigham AW , et al. Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet 2011; 12 (11) 745-755
  • 91 Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies for common human diseases. Nat Rev Genet 2011; 12 (8) 529-541
  • 92 Murphy SK, Yang H, Moylan CA , et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 2013; 145 (5) 1076-1087
  • 93 Greenbaum LE. From skin cells to hepatocytes: advances in application of iPS cell technology. J Clin Invest 2010; 120 (9) 3102-3105
  • 94 Pariente EA, Hamoud A, Goldfain D , et al. [Hepatitis caused by clometacin (Dupéran). Retrospective study of 30 cases. A model of autoimmune drug-induced hepatitis?]. Gastroenterol Clin Biol 1989; 13 (10) 769-774
  • 95 Hirata K, Takagi H, Yamamoto M , et al. Ticlopidine-induced hepatotoxicity is associated with specific human leukocyte antigen genomic subtypes in Japanese patients: a preliminary case-control study. Pharmacogenomics J 2008; 8 (1) 29-33
  • 96 Kurosaki M, Takagi H, Mori M. HLA-A33/B44/DR6 is highly related to intrahepatic cholestasis induced by tiopronin. Dig Dis Sci 2000; 45 (6) 1103-1108
  • 97 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 (7) 916-919
  • 98 Spraggs CF, Budde LR, Briley LP , et al. HLA-DQA1*02:01 is a major risk factor for lapatinib-induced hepatotoxicity in women with advanced breast cancer. J Clin Oncol 2011; 29 (6) 667-673
  • 99 Singer JB, Lewitzky S, Leroy E , et al. A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat Genet 2010; 42 (8) 711-714
  • 100 Phillips E, Bartlett JA, Sanne I , et al. Associations between HLA-DRB1*0102, HLA-B*5801, and hepatotoxicity during initiation of nevirapine-containing regimens in South Africa. J Acquir Immune Defic Syndr 2013; 62 (2) e55-e57
  • 101 Daly AK, Day CP. Genetic association studies in drug-induced liver injury. Drug Metab Rev 2012; 44 (1) 116-126