Drug-induced liver injury: recent advances in diagnosis and risk assessment

Gerd A Kullak-Ublick, Raul J Andrade, Michael Merz, Peter End, Andreas Benesic, Alexander L Gerbes, Guruprasad P Aithal, Gerd A Kullak-Ublick, Raul J Andrade, Michael Merz, Peter End, Andreas Benesic, Alexander L Gerbes, Guruprasad P Aithal

Abstract

Idiosyncratic drug-induced liver injury (IDILI) is a rare but potentially severe adverse drug reaction that should be considered in patients who develop laboratory criteria for liver injury secondary to the administration of a potentially hepatotoxic drug. Although currently used liver parameters are sensitive in detecting DILI, they are neither specific nor able to predict the patient's subsequent clinical course. Genetic risk assessment is useful mainly due to its high negative predictive value, with several human leucocyte antigen alleles being associated with DILI. New emerging biomarkers which could be useful in assessing DILI include total keratin18 (K18) and caspase-cleaved keratin18 (ccK18), macrophage colony-stimulating factor receptor 1, high mobility group box 1 and microRNA-122. From the numerous in vitro test systems that are available, monocyte-derived hepatocytes generated from patients with DILI show promise in identifying the DILI-causing agent from among a panel of coprescribed drugs. Several computer-based algorithms are available that rely on cumulative scores of known risk factors such as the administered dose or potential liabilities such as mitochondrial toxicity, inhibition of the bile salt export pump or the formation of reactive metabolites. A novel DILI cluster score is being developed which predicts DILI from multiple complimentary cluster and classification models using absorption-distribution-metabolism-elimination-related as well as physicochemical properties, diverse substructural descriptors and known structural liabilities. The provision of more advanced scientific and regulatory guidance for liver safety assessment will depend on validating the new diagnostic markers in the ongoing DILI registries, biobanks and public-private partnerships.

Keywords: ADVERSE DRUG REACTIONS; BILE ACID; DRUG INDUCED HEPATOTOXICITY; HEPATOBILIARY DISEASE; PHARMACOGENETICS.

Conflict of interest statement

Competing interests: AB and ALG have equity in the company MetaHeps GmbH. PE and MM are employees and GAK-U is a contractor of Novartis Pharma.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
Impact of idiosyncratic drug-induced liver injury (IDILI) on drug attrition. Pie charts showing the occurrence of liver test abnormalities in clinical trials with drugs withdrawn or stopped due to DILI. Blue: percentage of study participants with normal liver tests and Red: percentage of patients with possibly drug-related liver enzyme elevations.
Figure 2
Figure 2
Roussel-Uclaf Causality Assessment Method (RUCAM) diagnostic score.
Figure 3
Figure 3
Flow diagram of diagnostic workup of drug-induced liver injury (DILI). The phenotypes of liver injury are categorised according to the R value, defined as the ratio ALT/ULN:ALP/ULN. An R value of ≥5 indicates hepatocellular injury, ≤2 cholestatic injury and 2–5 mixed-type injury. ALP, alkaline phosphatase; ALT, alanine aminotransferase; CMV, cytomegalovirus; EBV, Epstein-Barr virus; HC, hepatocellular; HDSs, herbal and dietary supplements; OTC, over-the-counter drugs; PBC, primary biliary cirrhosis; PSC, primary sclerosing cholangitis; ULN, upper limit of normal.
Figure 4
Figure 4
(A) Example for a monocyte-derived hepatocyte-like (MH) cell test result from a patient with acute liver injury during treatment with sunitinib (for renal cell carcinoma), phenprocoumon (for atrial fibrillation) and metformin (for diabetes type II). MH cell toxicity is shown in a spiderweb graph. Sunitinib exerts marked toxicity in MH cells of this patient, whereas phenprocoumon and metformin do not show any effects. The red circle represents the individual cut-off for test positivity. (B) MH cell test results in 31 patients with idiosyncratic drug-induced liver injury (IDILI) and 23 patients with acute liver injury of other origin (non-DILI) using the drugs most likely to have caused liver injury in these cases. The MH cell test correctly identifies 29 of the 31 IDILI cases and shows no false-positive results. (C) MH cell test results using all drugs involved in the IDILI cases. Only four of the 84 comedications show positive results, suggesting that the MH cell test could be useful to identify the causative drug in complex IDILI cases. TWEEN, polyethylene glycol sorbitan monolaurate; ULN, upper limit of normal.
Figure 5
Figure 5
Interaction between drugs and human leucocyte antigen (HLA) molecule leading to an adaptive immune response. (A) Hapten hypothesis: drug–protein adducts (blue circles with red semicircle) released from dying hepatocytes are phagocytosed by antigen-presenting cells (APCs) and presented with major histocompatibility complex (MHC) II molecules. These hapten–carrier complexes bind to the peptide-binding groove on T cell receptors, leading to CD4+ cell activation and an effector T cell response. (B) Pharmacological interaction concept: drugs or metabolites can bind to HLA molecules directly and activate T cells. (C) Altered repertoire model: drug changes the shape and chemistry of the antigen-binding cleft, altering the repertoire of endogenous peptides that subsequently bind; the ‘altered self’ activates drug-specific T cells. (D) CD8+ cells recognise drug–protein adducts on the plasma membrane of hepatocytes when presented with MHC I molecules, leading to immunological destruction of hepatocytes.

References

    1. Lee WM. Drug-induced acute liver failure. Clin Liver Dis 2013;17:575–86, viii 10.1016/j.cld.2013.07.001
    1. Larrey D, Pageaux GP. Drug-induced acute liver failure. Eur J Gastroenterol Hepatol 2005;17:141–3. 10.1097/00042737-200502000-00002
    1. Fontana RJ, Hayashi PH, Gu J, et al. . Idiosyncratic drug-induced liver injury is associated with substantial morbidity and mortality within 6 months from onset. Gastroenterology 2014;147:96–108.e4. 10.1053/j.gastro.2014.03.045
    1. Sgro C, Clinard F, Ouazir K, et al. . Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology 2002;36:451–5. 10.1053/jhep.2002.34857
    1. Björnsson ES, Bergmann OM, Björnsson HK, et al. . Incidence, presentation, and outcomes in patients with drug-induced liver injury in the general population of Iceland. Gastroenterology 2013;144:1419–25, 25.e1–3; quiz e19–20 10.1053/j.gastro.2013.02.006
    1. Stevens JL, Baker TK. The future of drug safety testing: expanding the view and narrowing the focus. Drug Discov Today 2009;14:162–7. 10.1016/j.drudis.2008.11.009
    1. Lewis JH. ‘Hy's law,’ the ‘Rezulin Rule,’ and other predictors of severe drug-induced hepatotoxicity: putting risk-benefit into perspective. Pharmacoepidemiol Drug Saf 2006;15:221–9. 10.1002/pds.1209
    1. Nichols WG, Steel HM, Bonny T, et al. . Hepatotoxicity observed in clinical trials of aplaviroc (GW873140). Antimicrob Agents Chemother 2008;52:858–65. 10.1128/AAC.00821-07
    1. Lee WM, Larrey D, Olsson R, et al. . Hepatic findings in long-term clinical trials of ximelagatran. Drug Saf 2005;28:351–70. 10.2165/00002018-200528040-00006
    1. Schnitzer TJ, Burmester GR, Mysler E, et al. . Comparison of lumiracoxib with naproxen and ibuprofen in the Therapeutic Arthritis Research and Gastrointestinal Event Trial (TARGET), reduction in ulcer complications: randomised controlled trial. Lancet 2004;364:665–74. 10.1016/S0140-6736(04)16893-1
    1. Benza RL, Barst RJ, Galie N, et al. . Sitaxsentan for the treatment of pulmonary arterial hypertension: a 1-year, prospective, open-label observation of outcome and survival. Chest 2008;134:775–82. 10.1378/chest.07-0767
    1. Wysowski DK, Swartz L. Adverse drug event surveillance and drug withdrawals in the United States, 1969–2002: the importance of reporting suspected reactions. Arch Intern Med 2005;165:1363–9. 10.1001/archinte.165.12.1363
    1. 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;296:87–93. 10.1001/jama.296.1.87
    1. McGill MR, Jaeschke H. Metabolism and disposition of acetaminophen: recent advances in relation to hepatotoxicity and diagnosis. Pharm Res 2013;30:2174–87. 10.1007/s11095-013-1007-6
    1. McGill MR, Sharpe MR, Williams CD, et al. . The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J Clin Invest 2012;122:1574–83. 10.1172/JCI59755
    1. Reuben A, Koch DG, Lee WM, Acute Liver Failure Study Group. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology 2010;52:2065–76. 10.1002/hep.23937
    1. Yuan L, Kaplowitz N. Mechanisms of drug-induced liver injury. Clin Liver Dis 2013;17:507–18, vii 10.1016/j.cld.2013.07.002
    1. Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov 2005;4:489–99. 10.1038/nrd1750
    1. Russmann S, Jetter A, Kullak-Ublick GA. Pharmacogenetics of drug-induced liver injury. Hepatology 2010;52:748–61. 10.1002/hep.23720
    1. Aithal GP. Pharmacogenetic testing in idiosyncratic drug-induced liver injury: current role in clinical practice. Liver Int 2015;35:1801–8. 10.1111/liv.12836
    1. Lee KW, Chan SL. Hepatotoxicity of targeted therapy for cancer. Expert Opin Drug Metab Toxicol 2016;12:789–802. 10.1080/17425255.2016.1190831
    1. Takeda M, Okamoto I, Nakagawa K. Pooled safety analysis of EGFR-TKI treatment for EGFR mutation-positive non-small cell lung cancer. Lung Cancer 2015;88:74–9. 10.1016/j.lungcan.2015.01.026
    1. .
    1. Kullak-Ublick GA, Merz M, Griffel L, et al. . Liver safety assessment in special populations (hepatitis B, C, and oncology trials). Drug Saf 2014;37(Suppl 1):S57–62. 10.1007/s40264-014-0186-3
    1. Weiler S, Merz M, Kullak-Ublick GA. Drug-induced liver injury: the dawn of biomarkers? F1000Prime Rep 2015;7:34 10.12703/P7-34
    1. .
    1. .
    1. Chalasani NP, Hayashi PH, Bonkovsky HL, et al. . ACG Clinical Guideline: the diagnosis and management of idiosyncratic drug-induced liver injury. Am J Gastroenterol 2014;109:950–66; quiz 67 10.1038/ajg.2014.131
    1. Danan G, Teschke R. RUCAM in drug and herb induced liver injury: the update. Int J Mol Sci 2015;17:E14 10.3390/ijms17010014
    1. García-Cortés M, Stephens C, Lucena MI, et al. . Spanish Group for the Study of Drug-Induced Liver Disease (Grupo de Estudio para las Hepatopatías Asociadas a Medicamentos GEHAM). Causality assessment methods in drug induced liver injury: strengths and weaknesses. J Hepatol 2011;55:683–91. 10.1016/j.jhep.2011.02.007
    1. Hayashi PH. Drug-induced liver injury network causality assessment: criteria and experience in the United States. Int J Mol Sci 2016;17:201 10.3390/ijms17020201
    1. Rockey DC, Seeff LB, Rochon J, et al. . Causality assessment in drug-induced liver injury using a structured expert opinion process: comparison to the Roussel-Uclaf causality assessment method. Hepatology 2010;51:2117–26. 10.1002/hep.23577
    1. Teschke R, Schwarzenboeck A, Eickhoff A, et al. . Clinical and causality assessment in herbal hepatotoxicity. Expert Opin Drug Saf 2013;12:339–66. 10.1517/14740338.2013.774371
    1. Navarro VJ, Barnhart H, Bonkovsky HL, et al. . Liver injury from herbals and dietary supplements in the U.S. Drug-Induced Liver Injury Network. Hepatology 2014;60:1399–408. 10.1002/hep.27317
    1. Devarbhavi H, Raj S, Aradya VH, et al. . Drug-induced liver injury associated with Stevens-Johnson syndrome/toxic epidermal necrolysis: patient characteristics, causes, and outcome in 36 cases. Hepatology 2016;63:993–9. 10.1002/hep.28270
    1. Andrade RJ, Lucena MI, Fernández MC, et al. . Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 2005;129:512–21. 10.1016/j.gastro.2005.05.006
    1. 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;148:1340–52.e7. 10.1053/j.gastro.2015.03.006
    1. Andrade RJ, Robles M, Fernández-Castañer A, et al. . Assessment of drug-induced hepatotoxicity in clinical practice: a challenge for gastroenterologists. World J Gastroenterol 2007;13:329–40. 10.3748/wjg.v13.i3.329
    1. Davern TJ, Chalasani N, Fontana RJ, et al. . Acute hepatitis E infection accounts for some cases of suspected drug-induced liver injury. Gastroenterology 2011;141:1665–72.e1–9. 10.1053/j.gastro.2011.07.051
    1. Ortega-Alonso A, Stephens C, Lucena MI, et al. . Case characterization, clinical features and risk factors in drug-induced liver injury. Int J Mol Sci 2016;17:E714 10.3390/ijms17050714
    1. 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. 10.1038/clpt.2011.58
    1. Zimmerman HJ. The spectrum of hepatotoxicity. Perspect Biol Med 1968;12:135–61. 10.1353/pbm.1968.0004
    1. Björnsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology 2005;42:481–9. 10.1002/hep.20800
    1. Robles-Diaz M, Lucena MI, Kaplowitz N, et al. . Use of Hy's law and a new composite algorithm to predict acute liver failure in patients with drug-induced liver injury. Gastroenterology 2014;147:109–18.e5. 10.1053/j.gastro.2014.03.050
    1. de Boer YS, Kosinski AS, Urban TJ, et al. . Features of autoimmune hepatitis in patients with drug-induced liver injury. Clin Gastroenterol Hepatol 2017;15:103–112.e2. 10.1016/j.cgh.2016.05.043
    1. Lucena MI, Kaplowitz N, Hallal H, et al. . Recurrent drug-induced liver injury (DILI) with different drugs in the Spanish Registry: the dilemma of the relationship to autoimmune hepatitis. J Hepatol 2011;55:820–7. 10.1016/j.jhep.2010.12.041
    1. deLemos AS, Foureau DM, Jacobs C, et al. . Drug-induced liver injury with autoimmune features. Semin Liver Dis 2014;34:194–204. 10.1055/s-0034-1375959
    1. 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. 10.1002/hep.24481
    1. Foureau DM, Walling TL, Maddukuri V, et al. . Comparative analysis of portal hepatic infiltrating leucocytes in acute drug-induced liver injury, idiopathic autoimmune and viral hepatitis. Clin Exp Immunol 2015;180:40–51. 10.1111/cei.12558
    1. Andrade RJ, Robles M, Lucena MI. Rechallenge in drug-induced liver injury: the attractive hazard. Expert Opin Drug Saf 2009;8:709–14. 10.1517/14740330903397378
    1. Senior JR. Can rechallenge be done safely after mild or moderate drug-induced liver injury? Hepatology 2016;63:691–3. 10.1002/hep.28353
    1. 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;65:532–42. 10.1016/j.jhep.2016.05.003
    1. LeCluyse EL, Alexandre E, Hamilton GA, et al. . Isolation and culture of primary human hepatocytes. Methods Mol Biol 2005;290:207–29.
    1. Gómez-Lechón MJ, Lahoz A, Gombau L, et al. . In vitro evaluation of potential hepatotoxicity induced by drugs. Curr Pharm Des 2010;16:1963–77. 10.2174/138161210791208910
    1. Anson BD, Kolaja KL, Kamp TJ. Opportunities for use of human iPS cells in predictive toxicology. Clin Pharmacol Ther 2011;89:754–8. 10.1038/clpt.2011.9
    1. Lu J, Einhorn S, Venkatarangan L, et al. . Morphological and functional characterization and assessment of iPSC-derived hepatocytes for in vitro toxicity testing. Toxicol Sci 2015;147:39–54. 10.1093/toxsci/kfv117
    1. Atienzar FA, Blomme EA, Chen M, et al. . Key challenges and opportunities associated with the use of in vitro models to detect human DILI: integrated risk assessment and mitigation plans. Biomed Res Int 2016;2016: 9737920 10.1155/2016/9737920
    1. Godoy P, Hewitt NJ, Albrecht U, et al. . Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013;87:1315–530. 10.1007/s00204-013-1078-5
    1. Schadt S, Simon S, Kustermann S, et al. . Minimizing DILI risk in drug discovery—a screening tool for drug candidates. Toxicol In Vitro 2015;30:429–37. 10.1016/j.tiv.2015.09.019
    1. Tateno C, Miya F, Wake K, et al. . Morphological and microarray analyses of human hepatocytes from xenogeneic host livers. Lab Invest 2013;93:54–71. 10.1038/labinvest.2012.158
    1. Xu D, Wu M, Nishimura S, et al. . Chimeric TK-NOG mice: a predictive model for cholestatic human liver toxicity. J Pharmacol Exp Ther 2015;352:274–80. 10.1124/jpet.114.220798
    1. Boelsterli UA, Hsiao CJ. The heterozygous Sod2(+/-) mouse: modeling the mitochondrial role in drug toxicity. Drug Discov Today 2008;13:982–8. 10.1016/j.drudis.2008.08.002
    1. Lee YH, Chung MC, Lin Q, et al. . Troglitazone-induced hepatic mitochondrial proteome expression dynamics in heterozygous Sod2(+/-) mice: two-stage oxidative injury. Toxicol Appl Pharmacol 2008;231:43–51. 10.1016/j.taap.2008.03.025
    1. Benesic A, Rahm NL, Ernst S, et al. . Human monocyte-derived cells with individual hepatocyte characteristics: a novel tool for personalized in vitro studies. Lab Invest 2012;92:926–36. 10.1038/labinvest.2012.64
    1. Benesic A, Gerbes AL. Drug-induced liver injury and individual cell models. Dig Dis 2015;33:486–91. 10.1159/000374094
    1. Benesic A, Leitl A, Gerbes AL. Monocyte-derived hepatocyte-like cells for causality assessment of idiosyncratic drug-induced liver injury. Gut 2016;65:1555–63. 10.1136/gutjnl-2015-309528
    1. Danan G, Benichou C. Causality assessment of adverse reactions to drugs—I. A novel method based on the conclusions of international consensus meetings: application to drug-induced liver injuries. J Clin Epidemiol 1993;46:1323–30.
    1. Chen M, Vijay V, Shi Q, et al. . FDA-approved drug labeling for the study of drug-induced liver injury. Drug Discov Today 2011;16:697–703. 10.1016/j.drudis.2011.05.007
    1. Chen M, Suzuki A, Thakkar S, et al. . DILIrank: the largest reference drug list ranked by the risk for developing drug-induced liver injury in humans. Drug Discov Today 2016;21:648–53. 10.1016/j.drudis.2016.02.015
    1. Chen M, Borlak J, Tong W. High lipophilicity and high daily dose of oral medications are associated with significant risk for drug-induced liver injury. Hepatology 2013;58:388–96. 10.1002/hep.26208
    1. 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;42:744–50. 10.1124/dmd.113.056267
    1. Chen M, Borlak J, Tong W. A model to predict severity of drug-induced liver injury in humans. Hepatology 2016;64:931–40. 10.1002/hep.28678
    1. Aleo MD, Luo Y, Swiss R, et al. . Human drug-induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump. Hepatology 2014;60:1015–22. 10.1002/hep.27206
    1. Stieger B, Fattinger K, Madon J, et al. . Drug- and estrogen-induced cholestasis through inhibition of the hepatocellular bile salt export pump (Bsep) of rat liver. Gastroenterology 2000;118:422–30. 10.1016/S0016-5085(00)70224-1
    1. Morgan RE, Trauner M, van Staden CJ, et al. . Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development. Toxicol Sci 2010;118:485–500. 10.1093/toxsci/kfq269
    1. Dawson S, Stahl S, Paul N, et al. . In vitro inhibition of the bile salt export pump correlates with risk of cholestatic drug-induced liver injury in humans. Drug Metab Dispos 2012;40:130–8. 10.1124/dmd.111.040758
    1. Woodhead JL, Yang K, Siler SQ, et al. . Exploring BSEP inhibition-mediated toxicity with a mechanistic model of drug-induced liver injury. Front Pharmacol 2014;5:240 10.3389/fphar.2014.00240
    1. Longo DM, Yang Y, Watkins PB, et al. . Elucidating differences in the hepatotoxic potential of tolcapone and entacapone with DILIsym(®), a mechanistic model of drug-induced liver injury. CPT Pharmacometrics Syst Pharmacol 2016;5:31–9. 10.1002/psp4.12053
    1. Kuepfer L, Kerb R, Henney AM. Clinical translation in the virtual liver network. CPT Pharmacometrics Syst Pharmacol 2014;3:e127 10.1038/psp.2014.25
    1. Wang K, Zhang S, Marzolf B, et al. . Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc Natl Acad Sci USA 2009;106:4402–7. 10.1073/pnas.0813371106
    1. Starkey Lewis PJ, Dear J, Platt V, et al. . Circulating microRNAs as potential markers of human drug-induced liver injury. Hepatology 2011;54:1767–76. 10.1002/hep.24538
    1. Antoine DJ, Dear JW, Lewis PS, et al. . Mechanistic biomarkers provide early and sensitive detection of acetaminophen-induced acute liver injury at first presentation to hospital. Hepatology 2013;58:777–87. 10.1002/hep.26294
    1. Thulin P, Nordahl G, Gry M, et al. . Keratin-18 and microRNA-122 complement alanine aminotransferase as novel safety biomarkers for drug-induced liver injury in two human cohorts. Liver Int 2014;34:367–78. 10.1111/liv.12322
    1. Singhal R, Harrill AH, Menguy-Vacheron F, et al. . Benign elevations in serum aminotransferases and biomarkers of hepatotoxicity in healthy volunteers treated with cholestyramine. BMC Pharmacol Toxicol 2014;15:42 10.1186/2050-6511-15-42
    1. Starkey Lewis PJ, Merz M, Couttet P, et al. . Serum microRNA biomarkers for drug-induced liver injury. Clin Pharmacol Ther 2012;92:291–3. 10.1038/clpt.2012.101
    1. Antoine DJ, Jenkins RE, Dear JW, et al. . Molecular forms of HMGB1 and keratin-18 as mechanistic biomarkers for mode of cell death and prognosis during clinical acetaminophen hepatotoxicity. J Hepatol 2012;56:1070–9. 10.1016/j.jhep.2011.12.019
    1. Joka D, Wahl K, Moeller S, et al. . Prospective biopsy-controlled evaluation of cell death biomarkers for prediction of liver fibrosis and nonalcoholic steatohepatitis. Hepatology 2012;55:455–64. 10.1002/hep.24734
    1. Mikus M, Drobin K, Gry M, et al. . Elevated levels of circulating CDH5 and FABP1 in association with human drug-induced liver injury. Liver Int 2017;37:132–40. 10.1111/liv.13174
    1. Pelsers MM, Morovat A, Alexander GJ, et al. . Liver fatty acid-binding protein as a sensitive serum marker of acute hepatocellular damage in liver transplant recipients. Clin Chem 2002;48:2055–7.
    1. Akbal E, Köklü S, Koçak E, et al. . Liver fatty acid-binding protein is a diagnostic marker to detect liver injury due to chronic hepatitis C infection. Arch Med Res 2013;44:34–8. 10.1016/j.arcmed.2012.11.007
    1. Harrill AH, Roach J, Fier I, et al. . The effects of heparins on the liver: application of mechanistic serum biomarkers in a randomized study in healthy volunteers. Clin Pharmacol Ther 2012;92:214–20. 10.1038/clpt.2012.40
    1. Schomaker S, Warner R, Bock J, et al. . Assessment of emerging biomarkers of liver injury in human subjects. Toxicol Sci 2013;132:276–83. 10.1093/toxsci/kft009
    1. Beckett GJ, Foster GR, Hussey AJ, et al. . Plasma glutathione S-transferase and F protein are more sensitive than alanine aminotransferase as markers of paracetamol (acetaminophen)-induced liver damage. Clin Chem 1989;35:2186–9.
    1. Bailey WJ, Holder D, Patel H, et al. . A performance evaluation of three drug-induced liver injury biomarkers in the rat: alpha-glutathione S-transferase, arginase 1, and 4-hydroxyphenyl-pyruvate dioxygenase. Toxicol Sci 2012;130:229–44. 10.1093/toxsci/kfs243
    1. Ramaiah SK, Rittling S. Pathophysiological role of osteopontin in hepatic inflammation, toxicity, and cancer. Toxicol Sci 2008;103:4–13. 10.1093/toxsci/kfm246
    1. Arriazu E, Ge X, Leung TM, et al. . Signalling via the osteopontin and high mobility group box-1 axis drives the fibrogenic response to liver injury. Gut Published online first: 27 January 2016 10.1136/gutjnl-2015-310752
    1. Andersson U, Lindberg J, Wang S, et al. . A systems biology approach to understanding elevated serum alanine transaminase levels in a clinical trial with ximelagatran. Biomarkers 2009;14:572–86. 10.3109/13547500903261354
    1. McGill MR, Jaeschke H. Mechanistic biomarkers in acetaminophen-induced hepatotoxicity and acute liver failure: from preclinical models to patients. Expert Opin Drug Metab Toxicol 2014;10:1005–17. 10.1517/17425255.2014.920823
    1. Clarke JI, Dear JW, Antoine DJ. Recent advances in biomarkers and therapeutic interventions for hepatic drug safety—false dawn or new horizon? Expert Opin Drug Saf 2016;15:625–34. 10.1517/14740338.2016.1160057
    1. Woolbright BL, McGill MR, Staggs VS, et al. . Glycodeoxycholic acid levels as prognostic biomarker in acetaminophen-induced acute liver failure patients. Toxicol Sci 2014;142:436–44. 10.1093/toxsci/kfu195
    1. Schadt HS, Wolf A, Pognan F, et al. . Bile acids in drug induced liver injury: key players and surrogate markers. Clin Res Hepatol Gastroenterol 2016;40:257–66. 10.1016/j.clinre.2015.12.017
    1. Luo L, Schomaker S, Houle C, et al. . Evaluation of serum bile acid profiles as biomarkers of liver injury in rodents. Toxicol Sci 2014;137:12–25. 10.1093/toxsci/kft221
    1. De Berardinis V, Moulis C, Maurice M, et al. . Human microsomal epoxide hydrolase is the target of germander-induced autoantibodies on the surface of human hepatocytes. Mol Pharmacol 2000;58:542–51.
    1. Obermayer-Straub P, Strassburg CP, Manns MP. Target proteins in human autoimmunity: cytochromes P450 and UDP- glucuronosyltransferases. Can J Gastroenterol 2000;14:429–39. 10.1155/2000/910107
    1. Sutti S, Rigamonti C, Vidali M, et al. . CYP2E1 autoantibodies in liver diseases. Redox Biol 2014;3:72–8. 10.1016/j.redox.2014.11.004
    1. Metushi IG, Sanders C, Acute Liver Study Group, Lee WM, et al. . Detection of anti-isoniazid and anti-cytochrome P450 antibodies in patients with isoniazid-induced liver failure. Hepatology 2014;59:1084–93. 10.1002/hep.26564
    1. Gao H, Ruan JQ, Chen J, et al. . Blood pyrrole-protein adducts as a diagnostic and prognostic index in pyrrolizidine alkaloid-hepatic sinusoidal obstruction syndrome. Drug Des Devel Ther 2015;9:4861–8. 10.2147/DDDT.S87858
    1. Hindorff LA, Morales J, Junkins HA, et al. . A Catalog of Published Genome-Wide Association Studies.
    1. 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: 186–95. 10.1038/sj.tpj.6500458
    1. 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. 10.1038/ng.379
    1. Singer JB, Lewitzky S, Leroy E, et al. . A genome-wide study identifies HLA alleles associated with lumiracoxib-related liver injury. Nat Genet 2010;42:711–14. 10.1038/ng.632
    1. 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;29:667–73. 10.1200/JCO.2010.31.3197
    1. Parham LR, Briley LP, Li L, et al. . Comprehensive genome-wide evaluation of lapatinib-induced liver injury yields a single genetic signal centered on known risk allele HLA-DRB1*07:01. Pharmacogenomics J 2016;16:180–5. 10.1038/tpj.2015.40
    1. 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. 10.1053/j.gastro.2011.04.001
    1. Urban TJ, Shen Y, Stolz A, et al. . Limited contribution of common genetic variants to risk for liver injury due to a variety of drugs. Pharmacogenet Genomics 2012;22:784–95. 10.1097/FPC.0b013e3283589a76
    1. Nicoletti P, Werk AN, Sawle A, et al. . HLA-DRB1*16: 01-DQB1*05: 02 is a novel genetic risk factor for flupirtine-induced liver injury. Pharmacogenet Genomics 2016;26:218–24. 10.1097/FPC.0000000000000209
    1. Nicoletti P, Aithal GP, Bjornsson ES, et al. . Association of liver injury from specific drugs, or groups of drugs, with polymorphisms in HLA and other genes in a genome-wide association study. Gastroenterology 2016. doi:10.1053/j.gastro.2016.12.016 [Epub ahead of print 30 Dec 2016].10.1053/j.gastro.2016.12.016
    1. Urban TJ, Nicoletti P, Chalasani N, et al. . Minocycline hepatotoxicity: clinical characterization and identification of HLA-B 35:02 as a risk factor. Hepatology 2015;62(Suppl 1):1149A.
    1. Aithal GP, Grove JI. Genome-wide association studies in drug-induced liver injury: step change in understanding the pathogenesis. Semin Liver Dis 2015;35:421–31. 10.1055/s-0035-1567829
    1. 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;53:1049–53. 10.1016/j.jhep.2010.05.033
    1. Heap GA, Weedon MN, Bewshea CM, et al. . HLA-DQA1-HLA-DRB1 variants confer susceptibility to pancreatitis induced by thiopurine immunosuppressants. Nat Genet 2014;46:1131–4. 10.1038/ng.3093
    1. Grove JI, Aithal GP. Human leukocyte antigen genetic risk factors of drug-induced liver toxicology. Expert Opin Drug Metab Toxicol 2015;11:395–409. 10.1517/17425255.2015.992414
    1. Björnsson E, Aithal G. Immune-mediated drug-induced liver injury. In: Gershwin ME, Vierling JM, Manns MP, eds. Liver immunology. Springer International Publishing, 2014:401–12.
    1. Aithal GP, Daly AK. Preempting and preventing drug-induced liver injury. Nat Genet 2010;42:650–1. 10.1038/ng0810-650
    1. Tujios S, Fontana RJ. Mechanisms of drug-induced liver injury: from bedside to bench. Nat Rev Gastroenterol Hepatol 2011;8:202–11. 10.1038/nrgastro.2011.22

Source: PubMed

3
Abonnere