642
Views
69
CrossRef citations to date
0
Altmetric
Review

Biomarkers for drug-induced liver injury

, , &
Pages 225-234 | Published online: 10 Jan 2014

References

  • Chong CR, Sullivan DJ Jr. New uses for old drugs. Nature448, 645–646 (2007).
  • Hussaini SH, Farrington EA. Idiosyncratic drug-induced liver injury: an overview. Expert Opin. Drug Saf.6, 673–684 (2007).
  • Tarantino G, Pezzullo MG, Dario di Minno MN et al. Drug-induced liver injury due to ‘natural products’ used for weight loss: a case report. World J. Gastroenterol.15, 2414–2417 (2009).
  • Assis DN, Navarro VJ. Human drug hepatotoxicity: a contemporary clinical perspective. Expert Opin. Drug Metab. Toxicol.5, 463–473 (2009).
  • Kaplowitz N. Idiosyncratic drug hepatotoxicity. Nat. Rev. Drug Discov.4, 489–499 (2005).
  • Lee WM. Etiologies of acute liver failure. Semin. Liver Dis.28, 142–152 (2008).
  • Teschke R, Schwarzenboeck A, Hennermann KH. Causality assessment in hepatotoxicity by drugs and dietary supplements. Br. J. Clin. Pharmacol.66, 758–766 (2008).
  • Peters TS. Do preclinical testing strategies help predict human hepatotoxic potentials? Toxicol. Pathol.33, 146–154 (2005).
  • Dykens JA, Jamieson JD, Marroquin LD et al.In vitro assessment of mitochondrial dysfunction and cytotoxicity of nefazodone, trazodone, and buspirone. Toxicol. Sci.103, 335–345 (2008).
  • Tarantino G, Conca P, Basile V et al. A prospective study of acute drug-induced liver injury in patients suffering from non-alcoholic fatty liver disease. Hepatol. Res.37, 410–415 (2007).
  • Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints. preferred definitions and conceptual framework. Clin. Pharmacol. Ther.69, 89–95 (2001).
  • Senior JR. How can ‘Hy’s law’ help the clinician? Pharmacoepidemiol. Drug Saf.15, 235–239 (2006).
  • Ozer J, Ratner M, Shaw M et al. The current state of serum biomarkers of hepatotoxicity. Toxicology245, 194–205 (2008).
  • Yang RZ, Park S, Reagan WJ et al. Alanine aminotransferase isoenzymes. molecular cloning and quantitative analysis of tissue expression in rats and serum elevation in liver toxicity. Hepatology49, 598–607 (2009).
  • Edgar AD, Tomkiewicz C, Costet P et al. Fenofibrate modifies transaminase gene expression via a peroxisome proliferator activated receptor α-dependent pathway. Toxicol. Lett.98, 13–23 (1998).
  • Thulin P, Rafter I, Stockling K et al. PPARa regulates the hepatotoxic biomarker alanine aminotransferase (ALT1) gene expression in human hepatocytes. Toxicol. Appl. Pharmacol.231, 1–9 (2008).
  • Liu R, Pan X, Whitington PF. Increased hepatic expression is a major determinant of serum alanine aminotransferase elevation in mice with nonalcoholic steatohepatitis. Liver Int.29, 337–343 (2009).
  • Solter PF, Wollenberg GK, Huang X et al. Prolonged sublethal exposure to the protein phosphatase inhibitor microcystin-LR results in multiple dose-dependent hepatotoxic effects. Toxicol. Sci.44, 87–96 (1998).
  • Solter P, Liu Z, Guzman R. Decreased hepatic ALT synthesis is an outcome of subchronic microcystin-LR toxicity. Toxicol. Appl. Pharmacol.164, 216–220 (2000).
  • Miyazaki M, Rosenblum J, Kasahara Y et al. Determination of enzymatic source of alanine aminotransferase activity in serum from dogs with liver injury. J. Pharmacol. Toxicol. Methods60(3), 307–315 (2009).
  • Macchia T, Mancinelli R, Gentili S et al. Mitochondrial aspartate aminotransferase isoenzyme: a biochemical marker for the clinical management of alcoholics? Clin. Chim. Acta263, 79–96 (1997).
  • Musana KA, Yale SH, Abdulkarim AS. Tests of liver injury. Clin. Med. Res.2, 129–131 (2004).
  • Ramachandran R, Kakar S. Histological patterns in drug-induced liver disease. J. Clin. Pathol.62, 481–492 (2009).
  • Reust CE, Hall L. Clinical inquiries. What is the differential diagnosis of an elevated alkaline phosphatase (AP) level in an otherwise asymptomatic patient? J. Fam. Pract.50, 496–497 (2001).
  • Ohmori S, Shiraki K, Inoue H et al. Clinical characteristics and prognostic indicators of drug-induced fulminant hepatic failure. Hepatogastroenterology50, 1531–1534 (2003).
  • Senior JR. Monitoring for hepatotoxicity. What is the predictive value of liver ‘function’ tests? Clin. Pharmacol. Ther.85, 331–334 (2009).
  • Fabris L, Cadamuro M, Okolicsanyi L. The patient presenting with isolated hyperbilirubinemia. Dig. Liver Dis.41, 375–381 (2009).
  • Tarantino G. Could quantitative liver function tests gain wide acceptance among hepatologists? World J. Gastroenterol.15, 3457–3461 (2009).
  • Young TH, Tang HS, Chao YC et al. Quantitative rat liver function test by galactose single point method. Lab. Anim.42, 495–504 (2008).
  • Schmidt LE, Ott P, Tygstrup N. Galactose elimination capacity as a prognostic marker in patients with severe acetaminophen-induced hepatotoxicity: 10 years’ experience. Clin. Gastroenterol. Hepatol.2, 418–424 (2004).
  • Fontana RJ, Turgeon DK, Woolf TF et al. The caffeine breath test does not identify patients susceptible to tacrine hepatotoxicity. Hepatology23, 1429–1435 (1996).
  • Bopp SK, Lettieri T. Comparison of four different colorimetric and fluorometric cytotoxicity assays in a zebrafish liver cell line. BMC Pharmacol.8, 8 (2008).
  • Vinken M, Decrock E, De Vuyst E et al. Biochemical characterisation of an in vitro model of hepatocellular apoptotic cell death. Altern. Lab. Anim.37, 209–218 (2009).
  • O’Brien PJ, Slaughter MR, Polley SR et al. Advantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in rats. Lab. Anim.36, 313–321 (2002).
  • Halmes NC, Hinson JA, Martin BM et al. Glutamate dehydrogenase covalently binds to a reactive metabolite of acetaminophen. Chem. Res. Toxicol.9, 541–546 (1996).
  • Smith EL, Landon M, Piszkiewicz D et al. Bovine liver glutamate dehydrogenase. tentative amino acid sequence; identification of a reactive lysine; nitration of a specific tyrosine and loss of allosteric inhibition by guanosine triphosphate. Proc. Natl Acad. Sci. USA67, 724–730 (1970).
  • Zhang Y, Lu N, Gao Z. Hemin-H2O2-NO2- induced protein oxidation and tyrosine nitration are different from those of SIN-1: a study on glutamate dehydrogenase nitrative/oxidative modification. Int. J. Biochem. Cell. Biol.41, 907–915 (2009).
  • Banerjee S, Schmidt T, Fang J et al. Structural studies on ADP activation of mammalian glutamate dehydrogenase and the evolution of regulation. Biochemistry42, 3446–3456 (2003).
  • Lee WK, Shin S, Cho SS et al. Purification and characterization of glutamate dehydrogenase as another isoprotein binding to the membrane of rough endoplasmic reticulum. J. Cell. Biochem.76, 244–253 (1999).
  • James LP, Capparelli EV, Simpson PM et al. Acetaminophen-associated hepatic injury: evaluation of acetaminophen protein adducts in children and adolescents with acetaminophen overdose. Clin. Pharmacol. Ther.84, 684–690 (2008).
  • James LP, Letzig L, Simpson PM et al. Pharmacokinetics of acetaminophen–protein adducts in adults with acetaminophen overdose and acute liver failure. Drug Metab. Dispos.37, 1779–1784 (2009).
  • Wade LT, Kenna JG, Caldwell J. Immunochemical identification of mouse hepatic protein adducts derived from the nonsteroidal anti-inflammatory drugs diclofenac, sulindac and ibuprofen. Chem. Res. Toxicol.10, 546–555 (1997).
  • Heijne WH, Stierum RH, Leeman WR et al. The introduction of toxicogenomics; potential new markers of hepatotoxicity. Cancer Biomark.1, 41–57 (2005).
  • Saha B, Nandi D. Farnesyltransferase inhibitors reduce ras activation and ameliorate acetaminophen-induced liver injury in mice. Hepatology50(5), 1547–1557 (2009).
  • Rutherford AE, Hynan LS, Borges CB et al. Serum apoptosis markers in acute liver failure. a pilot study. Clin. Gastroenterol. Hepatol.5, 1477–1483 (2007).
  • Tagami A, Ohnishi H, Hughes RD. Increased serum soluble Fas in patients with acute liver failure due to paracetamol overdose. Hepatogastroenterology50, 742–745 (2003).
  • Singhal S, Chakravarty A, Das BC et al. Tumour necrosis factor-a and soluble Fas ligand as biomarkers in non-acetaminophen-induced acute liver failure. Biomarkers14, 347–353 (2009).
  • Carr DF, Alfirevic A, Tugwood JD et al. Molecular and genetic association of interleukin-6 in tacrine-induced hepatotoxicity. Pharmacogenet. Genomics17, 961–972 (2007).
  • Aithal GP, Ramsay L, Daly AK et al. Hepatic adducts, circulating antibodies, and cytokine polymorphisms in patients with diclofenac hepatotoxicity. Hepatology39, 1430–1440 (2004).
  • Pachkoria K, Lucena MI, Crespo E et al. Analysis of IL-10, IL-4 and TNF-α polymorphisms in drug-induced liver injury (DILI) and its outcome. J. Hepatol.49, 107–114 (2008).
  • Pham BN, Bemuau J, Durand F et al. Eotaxin expression and eosinophil infiltrate in the liver of patients with drug-induced liver disease. J. Hepatol.34, 537–547 (2001).
  • Tarantino G, Di Minno MN, Capone D. Drug-induced liver injury. Is it somehow foreseeable? World J. Gastroenterol.15, 2817–2833 (2009).
  • Grismer LE, Gill SA, Harris MD. Liver biopsy in psoriatic arthritis to detect methotrexate hepatotoxicity. J. Clin. Rheumatol.7, 224–227 (2001).
  • Blomme EA, Yang Y, Waring JF. Use of toxicogenomics to understand mechanisms of drug-induced hepatotoxicity during drug discovery and development. Toxicol. Lett.186, 22–31 (2009).
  • Liguori MJ, Anderson MG, Bukofzer S et al. Microarray analysis in human hepatocytes suggests a mechanism for hepatotoxicity induced by trovafloxacin. Hepatology41, 177–186 (2005).
  • Natsoulis G, Pearson CI, Gollub J et al. The liver pharmacological and xenobiotic gene response repertoire. Mol. Syst. Biol.4, 175 (2008).
  • McMillian M, Nie AY, Parker JB et al. A gene expression signature for oxidant stress/reactive metabolites in rat liver. Biochem. Pharmacol.68, 2249–2261 (2004).
  • McMillian M, Nie AY, Parker JB et al. Inverse gene expression patterns for macrophage activating hepatotoxicants and peroxisome proliferators in rat liver. Biochem. Pharmacol.67, 2141–2165 (2004).
  • McMillian M, Nie A, Parker JB et al. Drug-induced oxidative stress in rat liver from a toxicogenomics perspective. Toxicol. Appl. Pharmacol.207, 171–178 (2005).
  • Yuan L, Kaplowitz N. Glutathione in liver diseases and hepatotoxicity. Mol. Aspects Med.30, 29–41 (2009).
  • Kiyosawa N, Ito K, Sakuma K et al. Evaluation of glutathione deficiency in rat livers by microarray analysis. Biochem. Pharmacol.68, 1465–1475 (2004).
  • Kiyosawa N, Uehara T, Gao W et al. Identification of glutathione depletion-responsive genes using phorone-treated rat liver. J. Toxicol. Sci.32, 469–486 (2007).
  • Reasor MJ, Kacew S. Drug-induced phospholipidosis: are there functional consequences? Exp. Biol. Med. (Maywood)226, 825–830 (2001).
  • Anderson N, Borlak J. Drug-induced phospholipidosis. FEBS Lett.580, 5533–5540 (2006).
  • Hirode M, Ono A, Miyagishima T et al. Gene expression profiling in rat liver treated with compounds inducing phospholipidosis. Toxicol. Appl. Pharmacol.229, 290–299 (2008).
  • Sawada H, Takami K, Asahi S. A toxicogenomic approach to drug-induced phospholipidosis: analysis of its induction mechanism and establishment of a novel in vitro screening system. Toxicol. Sci.83, 282–292 (2005).
  • Atienzar F, Gerets H, Dufrane S et al. Determination of phospholipidosis potential based on gene expression analysis in HepG2 cells. Toxicol. Sci.96, 101–114 (2007).
  • Ganey PE, Luyendyk JP, Newport SW et al. Role of the coagulation system in acetaminophen-induced hepatotoxicity in mice. Hepatology46, 1177–1186 (2007).
  • Hirode M, Omura K, Kiyosawa N et al. Gene expression profiling in rat liver treated with various hepatotoxic-compounds inducing coagulopathy. J. Toxicol. Sci.34, 281–293 (2009).
  • Bushel PR, Heinloth AN, Li J et al. Blood gene expression signatures predict exposure levels. Proc. Natl Acad. Sci. USA104, 18211–18216 (2007).
  • Umbright C, Sellamuthu R, Li S et al. Blood gene expression markers to detect and distinguish target organ toxicity. Mol. Cell. Biochem.335(1–2), 223–234 (2009).
  • Boelsterli UA, Lim PL. Mitochondrial abnormalities – a link to idiosyncratic drug hepatotoxicity? Toxicol. Appl. Pharmacol.220, 92–107 (2007).
  • Lammert C, Einarsson S, Saha C et al. Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology47, 2003–2009 (2008).
  • Senior JR. What is idiosyncratic hepatotoxicity? What is it not? Hepatology47, 1813–1815 (2008).
  • Uetrecht J. Idiosyncratic drug reactions. current understanding. Annu. Rev. Pharmacol. Toxicol.47, 513–539 (2007).
  • Zou W, Beggs KM, Sparkenbaugh EM et al. Sulindac metabolism and synergy with TNF in a drug-inflammation interaction model of idiosyncratic liver injury. J. Pharmacol. Exp. Ther.331(1), 114–121 (2009).
  • Kashimshetty R, Desai VG, Kale VM et al. Underlying mitochondrial dysfunction triggers flutamide-induced oxidative liver injury in a mouse model of idiosyncratic drug toxicity. Toxicol. Appl. Pharmacol.238, 150–159 (2009).
  • Gao J, Ann Garulacan L, Storm SM et al. Identification of in vitro protein biomarkers of idiosyncratic liver toxicity. Toxicol. In Vitro18, 533–541 (2004).
  • Mendrick DL. Toxicogenomics and classic toxicology. How to improve prediction and mechanistic understanding of human toxicity. Methods Mol. Biol.460, 1–22 (2008).
  • Mendrick DL. Genomic and genetic biomarkers of toxicity. Toxicology245, 175–181 (2008).
  • Andrade RJ, Robles M, Ulzurrun E et al. Drug-induced liver injury: insights from genetic studies. Pharmacogenomics10, 1467–1487 (2009).
  • Russmann S, Kullak-Ublick GA, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr. Med. Chem.16, 3041–3053 (2009).
  • 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.41, 816–819 (2009).
  • Bartels CL, Tsongalis GJ. MicroRNAs: novel biomarkers for human cancer. Clin. Chem.55, 623–631 (2009).
  • Cortez MA, Calin GA. MicroRNA identification in plasma and serum: a new tool to diagnose and monitor diseases. Expert Opin. Biol. Ther.9, 703–711 (2009).
  • Miyamoto M, Yanai M, Ookubo S et al. Detection of cell-free, liver-specific mRNAs in peripheral blood from rats with hepatotoxicity: a potential toxicological biomarker for safety evaluation. Toxicol. Sci.106, 538–545 (2008).
  • Wang K, Zhang S, Marzolf B et al. Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc. Natl Acad. Sci. USA106, 4402–4407 (2009).
  • Kudo Y, Ochi T, Shimada H et al. Utility of plasma circulating mRNA as a marker to detect hepatic injury. J. Vet. Med. Sci.70, 993–995 (2008).
  • Laterza OF, Lim L, Garrett-Engele PW et al. Plasma microRNAs as diagnostically sensitive and specific biomarkers of tissue injury. Clin. Chem.55(11), 1977–1983 (2009).
  • Soga T, Baran R, Suematsu M et al. Differential metabolomics reveals ophthalmic acid as an oxidative stress biomarker indicating hepatic glutathione consumption. J. Biol. Chem.281, 16768–16776 (2006).
  • Lee MS, Jung BH, Chung BC et al. Metabolomics study with gas chromatography-mass spectrometry for predicting valproic acid-induced hepatotoxicity and discovery of novel biomarkers in rat urine. Int. J. Toxicol.28(5), 392–404 (2009).
  • Amacher DE, Adler R, Herath A et al. Use of proteomic methods to identify serum biomarkers associated with rat liver toxicity or hypertrophy. Clin. Chem.51, 1796–1803 (2005).
  • Goodsaid F, Frueh F. Biomarker qualification pilot process at the US Food and Drug Administration. AAPS J.9, E105–E108 (2007).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.