Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-cnmwb Total loading time: 0 Render date: 2024-07-22T10:03:22.187Z Has data issue: false hasContentIssue false

4 - Predicting drug-induced liver injury: safer patients or safer drugs?

from I - SPECIFIC AREAS OF PREDICTIVE TOXICOLOGY

Published online by Cambridge University Press:  06 December 2010

Jinghai J. Xu
Affiliation:
Merck Research Laboratory, New Jersey
Laszlo Urban
Affiliation:
Novartis Institutes for Biomedical Research, Massachusetts
Get access

Summary

INTRODUCTION: THE PROBLEM OF DRUG-INDUCED LIVER INJURY

Liver injury or hepatotoxicity caused by medications has been a major challenge for health care professionals and drug developers alike. In the United States alone, drug-induced liver injury (DILI) is responsible for 5 percent of all hospital admissions and 50 percent of all acute liver failures. DILI is the most common reason for black-box warnings among drugs previously approved for marketing, as well as drug withdrawals after regulatory approval for marketing. DILI is also a leading reason for nonapproval decisions by the Food and Drug Administration (FDA) in the United States, and a major reason why drugs fail in clinical and preclinical development.

Drugs for which development, approval, or clinical usage were significantly impacted due to hepatotoxicity include troglitazone, bromfenac, tienilic acid, temafloxacin, nomifensin, perhexiline, ibufenac, benoxaprofen, zileutin, trovafloxacin, tolcapone, felbamate, iproniazid, ticrynafen, labetalol, alpidem, ebrotidine, dilevalol, tasosartan, telithromycin, and ximelagatran. Several textbooks have been published on the topic of DILI (e.g., References 5, 6). There is also an annual summary of drugs associated with hepatotoxicity, which provides excellent updates on this topic.

Regulators of the pharmaceutical industry have made it a priority to provide clinical and preclinical guidance to improve detection of DILI. Databases of potentially hepatotoxic drugs have been established, interagency collaborations aimed at better prediction of DILI are underway.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

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

Ostapowicz, G, Fontana, RJ, Schiodt, FV, et al. 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.CrossRefGoogle ScholarPubMed
Temple, RJ. Hepatotoxicity through the years impact on the FDA. Paper presented at Drug-Induced Liver Toxicity, Silver Spring, Maryland; 2001.
Schuster, D, Laggner, C, Langer, T. Why drugs fail – A study on side effects in new chemical entities. Curr Pharm Des. 2005;11(27):3545–3559.CrossRefGoogle ScholarPubMed
Peters, TS. Do preclinical testing strategies help predict human hepatotoxic potentials?Toxicol Pathol. 2005;33(1):146–154.CrossRefGoogle ScholarPubMed
Zimmerman, H. Hepatotoxicity: The Adverse Effects of Drugs and Other Chemicals on the Liver. 2nd ed. Philadelphia: Lippincott Williams & Wilkins; 1999.Google Scholar
Stricker, BHC, ed. Drug-Induced Hepatic Injury. 2nd ed. Amsterdam: Elsevier; 1992.
Norris, W, Paredes, AH, Lewis, JH. Drug-induced liver injury in 2007. Curr Opin Gastroenterol. 2008;24(3):287–297.CrossRefGoogle ScholarPubMed
,FDA. Drug-Induced Liver Injury: Premarketing Clinical Evaluation. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM174090.pdf. Accessed 2010.
,EMEA. Non-clinical Guideline on Drug-Induced Hepatotoxicity. http://www.ema.europa.eu/pdfs/human/swp/15011506en.pdf. Accessed 2010.
Guo, JJ, Wigle, PR, Lammers, K, Vu, O. Comparison of potentially hepatotoxic drugs among major US drug compendia. Res Social Adm Pharm. 2005;1(3):460–479.CrossRefGoogle ScholarPubMed
Fontana, RJ, Watkins, PB, Bonkovsky, HL, et al. Drug-induced liver injury network (DILIN) prospective study: Rationale, design and conduct. Drug Saf. 2009;32(1):55–68.CrossRefGoogle ScholarPubMed
Rochon, J, Protiva, P, Seeff, LB, et al. Reliability of the Roussel Uclaf Causality Assessment Method for assessing causality in drug-induced liver injury. Hepatology. 2008;48(4):1175–1183.CrossRefGoogle ScholarPubMed
,DILIN. DILIN Overview. https://dilin.dcri.duke.edu/for-researchers/. Accessed 2010.
Kaplowitz, N. Idiosyncratic drug hepatotoxicity. Nat Rev Drug Discov. 2005; 4(6):489–499.CrossRefGoogle ScholarPubMed
Wynne, H. Drug metabolism and ageing. J Br Menopause Soc. 2005;11(2):51–56.CrossRefGoogle ScholarPubMed
Muhlberg, W, Platt, D. Age-dependent changes of the kidneys: Pharmacological implications. Gerontology. 1999;45(5):243–253.CrossRefGoogle ScholarPubMed
Cusack, BJ. Pharmacokinetics in older persons. Am J Geriatr Pharmacother. 2004;2(4):274–302.CrossRefGoogle ScholarPubMed
Reis, M, Aamo, T, Spigset, O, Ahlner, J. Serum concentrations of antidepressant drugs in a naturalistic setting: Compilation based on a large therapeutic drug monitoring database. Ther Drug Monit. 2009;31(1):42–56.CrossRefGoogle Scholar
Schwartz, JB. The influence of sex on pharmacokinetics. Clin Pharmacokinet. 2003;42(2):107–121.CrossRefGoogle ScholarPubMed
Feng, B, Xu, JJ, Bi, YA, et al. Role of hepatic transporters in the disposition and hepatotoxicity of a HER2 tyrosine kinase inhibitor CP-724,714. Toxicol Sci. 2009;108(2):492–500.CrossRefGoogle ScholarPubMed
Fukudo, M, Yano, I, Yoshimura, A, et al. Impact of MDR1 and CYP3A5 on the oral clearance of tacrolimus and tacrolimus-related renal dysfunction in adult living-donor liver transplant patients. Pharmacogenet Genomics. 2008;18(5):413–423.CrossRefGoogle ScholarPubMed
Fabbiani, M, Di Giambenedetto, S, Bracciale, L, et al. Pharmacokinetic variability of antiretroviral drugs and correlation with virological outcome: 2 years of experience in routine clinical practice. J Antimicrob Chemother. 2009;64(1):109–117.CrossRefGoogle ScholarPubMed
Reis, M, Cherma, MD, Carlsson, B, Bengtsson, F. Therapeutic drug monitoring of escitalopram in an outpatient setting. Ther Drug Monit. 2007;29(6):758–766.CrossRefGoogle Scholar
Findling, RL, Reed, MD, Myers, C, et al. Paroxetine pharmacokinetics in depressed children and adolescents. J Am Acad Child Adolesc Psychiatry. 1999;38(8): 952–959.CrossRefGoogle ScholarPubMed
Ebbesen, J, Buajordet, I, Erikssen, J, et al. Drug-related deaths in a department of internal medicine. Arch Intern Med. 2001;161(19):2317–2323.CrossRefGoogle Scholar
Garcia-Compean, D, Jaquez-Quintana, JO, Maldonado-Garza, H. Hepatogenous diabetes. Current views of an ancient problem. Ann Hepatol. 2009;8(1):13–20.Google ScholarPubMed
Wang, T, Shankar, K, Ronis, MJ, Mehendale, HM. Mechanisms and outcomes of drug- and toxicant-induced liver toxicity in diabetes. Crit Rev Toxicol. 2007;37(5):413–459.CrossRefGoogle ScholarPubMed
Douglas, RM, Haddad, GG. Can O2 dysregulation induce premature aging?Physiology (Bethesda). 2008;23:333–349.Google ScholarPubMed
Pialoux, V, Hanly, PJ, Foster, GE, et al. Effects of exposure to intermittent hypoxia on oxidative stress and acute hypoxic ventilatory response in humans. Am J Respir Crit Care Med. 2009;180(10):1002–1009.CrossRefGoogle ScholarPubMed
Savransky, V, Nanayakkara, A, Vivero, A, et al. Chronic intermittent hypoxia predisposes to liver injury. Hepatology. 2007;45(4):1007–1013.CrossRefGoogle ScholarPubMed
Savransky, V, Reinke, C, Jun, J, et al. Chronic intermittent hypoxia and acetaminophen induce synergistic liver injury in mice. Exp Physiol. 2009;94(2):228–239.CrossRefGoogle ScholarPubMed
Dixit, R, Boelsterli, UA. Healthy animals and animal models of human disease(s) in safety assessment of human pharmaceuticals, including therapeutic antibodies. Drug Discov Today. 2007;12(7–8):336–342.CrossRefGoogle ScholarPubMed
Wu, D, Cederbaum, AI. Oxidative stress and alcoholic liver disease. Semin Liver Dis. 2009;29(2):141–154.CrossRefGoogle ScholarPubMed
Donthamsetty, S, Bhave, VS, Mitra, MS, Latendresse, JR, Mehendale, HM. Nonalcoholic fatty liver sensitizes rats to carbon tetrachloride hepatotoxicity. Hepatology. 2007;45(2):391–403.CrossRefGoogle ScholarPubMed
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. 2007;37(6):410–415.CrossRefGoogle ScholarPubMed
Musso, G, Gambino, R, Durazzo, M, et al. Adipokines in NASH: Postprandial lipid metabolism as a link between adiponectin and liver disease. Hepatology. 2005;42(5):1175–1183.CrossRefGoogle ScholarPubMed
Musso, G, Gambino, R, Pacini, G, Michieli, F, Cassader, M. Prolonged saturated fat-induced, glucose-dependent insulinotropic polypeptide elevation is associated with adipokine imbalance and liver injury in nonalcoholic steatohepatitis: Dysregulated enteroadipocyte axis as a novel feature of fatty liver. Am J Clin Nutr. 2009;89(2):558–567.CrossRefGoogle ScholarPubMed
Liu, J, Zhou, ZX, Zhang, W, Bell, MW, Waalkes, MP. Changes in hepatic gene expression in response to hepatoprotective levels of zinc. Liver Int. 2009;29(8):1222–1229.CrossRefGoogle ScholarPubMed
Li, X, Chen, H, Epstein, PN. Metallothionein protects islets from hypoxia and extends islet graft survival by scavenging most kinds of reactive oxygen species. J Biol Chem. 2004;279(1):765–771.CrossRefGoogle ScholarPubMed
Edling, Y, Sivertsson, LK, Butura, A, Ingelman-Sundberg, M, Ek, M. Increased sensitivity for troglitazone-induced cytotoxicity using a human in vitro co-culture model. Toxicol In Vitro. 2009;23(7):1387–1395.CrossRefGoogle ScholarPubMed
Cosgrove, BD, King, BM, Hasan, MA, et al. Synergistic drug-cytokine induction of hepatocellular death as an in vitro approach for the study of inflammation-associated idiosyncratic drug hepatotoxicity. Toxicol Appl Pharmacol. 2009;237(3):317–330.CrossRefGoogle Scholar
Chang, CY, Schiano, TD. Review article: Drug hepatotoxicity. Aliment Pharmacol Ther. 2007;25(10):1135–1151.CrossRefGoogle ScholarPubMed
El-Sherbiny, GA, Taye, A, Abdel-Raheem, IT. Role of ursodeoxycholic acid in prevention of hepatotoxicity caused by amoxicillin-clavulanic acid in rats. Ann Hepatol. 2009;8(2):134–140.Google ScholarPubMed
Upreti, VV, Eddington, ND, Moon, KH, Song, BJ, Lee, IJ. Drug interaction between ethanol and 3,4-methylenedioxymethamphetamine (“ecstasy”). Toxicol Lett. 2009;188(2):167–172.CrossRefGoogle Scholar
Shaw, PJ, Hopfensperger, MJ, Ganey, PE, Roth, RA. Lipopolysaccharide and trovafloxacin coexposure in mice causes idiosyncrasy-like liver injury dependent on tumor necrosis factor-alpha. Toxicol Sci. Nov 2007;100(1):259–266.CrossRefGoogle ScholarPubMed
Verhelst, X, Burvenich, P, Sassenbroeck, D, Gabriel, C, Lootens, M, Baert, D. Acute hepatitis after treatment for hair loss with oral green tea extracts (Camellia sinensis). Acta Gastroenterol Belg. 2009;72(2):262–264.Google Scholar
Stickel, F, Droz, S, Patsenker, E, Bogli-Stuber, K, Aebi, B, Leib, SL. Severe hepatotoxicity following ingestion of Herbalife nutritional supplements contaminated with Bacillus subtilis. J Hepatol. 2009;50(1):111–117.CrossRefGoogle ScholarPubMed
Foti, RS, Dickmann, LJ, Davis, JA, et al. Metabolism and related human risk factors for hepatic damage by usnic acid containing nutritional supplements. Xenobiotica. 2008;38(3):264–280.CrossRefGoogle ScholarPubMed
Uppal, H, Saini, SP, Moschetta, A, et al. Activation of LXRs prevents bile acid toxicity and cholestasis in female mice. Hepatology. 2007;45(2):422–432.CrossRefGoogle ScholarPubMed
Yamamoto, Y, Moore, R, Hess, HA, et al. Estrogen receptor alpha mediates 17alpha-ethynylestradiol causing hepatotoxicity. J Biol Chem. 2006;281(24): 16625–16631.CrossRefGoogle ScholarPubMed
Osburn, WO, Kensler, TW. Nrf2 signaling: An adaptive response pathway for protection against environmental toxic insults. Mutat Res. 2008;659(1–2):31–39.CrossRefGoogle ScholarPubMed
Lucena, MI, Andrade, RJ, Martinez, C, et al. Glutathione S-transferase m1 and t1 null genotypes increase susceptibility to idiosyncratic drug-induced liver injury. Hepatology. 2008;48(2):588–596.CrossRefGoogle ScholarPubMed
Marcos, M, Pastor, I, Gonzalez-Sarmiento, R, Laso, FJ. A functional polymorphism of the NFKB1 gene increases the risk for alcoholic liver cirrhosis in patients with alcohol dependence. Alcohol Clin Exp Res. 2009;33(11):1857–1862.CrossRefGoogle ScholarPubMed
Shah, RR. Can pharmacogenetics help rescue drugs withdrawn from the market?Pharmacogenomics. 2006;7(6):889–908.CrossRefGoogle Scholar
Gardiner, SJ, Begg, EJ. Pharmacogenetics, drug-metabolizing enzymes, and clinical practice. Pharmacol Rev. Sep 2006;58(3):521–590.CrossRefGoogle ScholarPubMed
Gardiner, SJ, Begg, EJ. Pharmacogenetic testing for drug metabolizing enzymes: Is it happening in practice?Pharmacogenet Genomics. 2005;15(5):365–369.CrossRefGoogle ScholarPubMed
Barclay, ML, Sawyers, SM, Begg, EJ, et al. Correlation of CYP2D6 genotype with perhexiline phenotypic metabolizer status. Pharmacogenetics. 2003;13(10): 627–632.CrossRefGoogle ScholarPubMed
Gaedigk, A, Simon, SD, Pearce, RE, Bradford, LD, Kennedy, MJ, Leeder, JS. The CYP2D6 activity score: Translating genotype information into a qualitative measure of phenotype. Clin Pharmacol Ther. 2008;83(2):234–242.CrossRefGoogle ScholarPubMed
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(4):221–229.CrossRefGoogle Scholar
Kaplowitz, N. Rules and laws of drug hepatotoxicity. Pharmacoepidemiol Drug Saf. 2006;15(4):231–233.CrossRefGoogle ScholarPubMed
Ozer, J, Ratner, M, Shaw, M, Bailey, W, Schomaker, S. The current state of serum biomarkers of hepatotoxicity. Toxicology. 2008;245(3):194–205.CrossRefGoogle ScholarPubMed
Lee, WM. Drug-induced hepatotoxicity. N Engl J Med. 2003;349(5):474–485.CrossRefGoogle ScholarPubMed
Armitage, P, Doll, R. A two-stage theory of carcinogenesis in relation to the age distribution of human cancer. Br J Cancer. 1957;11(2):161–169.CrossRefGoogle ScholarPubMed
Emmelot, P, Scherer, E. Multi-hit kinetics of tumor formation, with special reference to experimental liver and human lung carcinogenesis and some general conclusions. Cancer Res. 1977;37(6):1702–1708.Google Scholar
Ashley, DJ. The two “hit” and multiple “hit” theories of carcinogenesis. Br J Cancer. 1969;23(2):313–328.CrossRefGoogle ScholarPubMed
Pessayre, D, Berson, A, Fromenty, B, Mansouri, A. Mitochondria in steatohepatitis. Semin Liver Dis. 2001;21(1):57–69.CrossRefGoogle ScholarPubMed
Begriche, K, Igoudjil, A, Pessayre, D, Fromenty, B. Mitochondrial dysfunction in NASH: Causes, consequences and possible means to prevent it. Mitochondrion. 2006;6(1):1–28.CrossRefGoogle Scholar
Zou, W, Beggs, KM, Sparkenbaugh, EM, et al. Sulindac metabolism and synergy with tumor necrosis factor-alpha in a drug-inflammation interaction model of idiosyncratic liver injury. J Pharmacol Exp Ther. 2009;331(1):114–121.CrossRefGoogle Scholar
Shaw, PJ, Ganey, PE, Roth, RA. Trovafloxacin enhances the inflammatory response to a Gram-negative or a Gram-positive bacterial stimulus, resulting in neutrophil-dependent liver injury in mice. J Pharmacol Exp Ther. 2009;330(1):72–78.CrossRefGoogle ScholarPubMed
Shaw, PJ, Ditewig, AC, Waring, JF, et al. Coexposure of mice to trovafloxacin and lipopolysaccharide, a model of idiosyncratic hepatotoxicity, results in a unique gene expression profile and interferon gamma-dependent liver injury. Toxicol Sci. 2009;107(1):270–280.CrossRefGoogle Scholar
Deng, X, Liguori, MJ, Sparkenbaugh, EM, et al. Gene expression profiles in livers from diclofenac-treated rats reveal intestinal bacteria-dependent and -independent pathways associated with liver injury. J Pharmacol Exp Ther. 2008;327(3): 634–644.CrossRefGoogle ScholarPubMed
Lee, YH, Boelsterli, UA, Lin, Q, Chung, MC. Proteomics profiling of hepatic mitochondria in heterozygous Sod2+/- mice, an animal model of discreet mitochondrial oxidative stress. Proteomics. 2008;8(3):555–568.CrossRefGoogle Scholar
Lee, YH, Chung, MC, Lin, Q, Boelsterli, UA. Troglitazone-induced hepatic mitochondrial proteome expression dynamics in heterozygous Sod2(+/-) mice: Two-stage oxidative injury. Toxicol Appl Pharmacol. 2008;231(1):43–51.CrossRefGoogle ScholarPubMed
Ong, MM, Wang, AS, Leow, KY, Khoo, YM, Boelsterli, UA. Nimesulide-induced hepatic mitochondrial injury in heterozygous Sod2(+/-) mice. Free Radic Biol Med. 2006;40(3):420–429.CrossRefGoogle ScholarPubMed
Boelsterli, UA. Animal models of human disease in drug safety assessment. J Toxicol Sci. 2003;28(3):109–121.CrossRefGoogle ScholarPubMed
James, LP, Mayeux, PR, Hinson, JA. Acetaminophen-induced hepatotoxicity. Drug Metab Dispos. 2003;31(12):1499–1506.CrossRefGoogle ScholarPubMed
Reid, AB, Kurten, RC, McCullough, SS, Brock, RW, Hinson, JA. Mechanisms of acetaminophen-induced hepatotoxicity: Role of oxidative stress and mitochondrial permeability transition in freshly isolated mouse hepatocytes. J Pharmacol Exp Ther. 2005;312(2):509–516.CrossRefGoogle ScholarPubMed
Hanawa, N, Shinohara, M, Saberi, B, Gaarde, WA, Han, D, Kaplowitz, N. Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminophen-induced liver injury. J Biol Chem. 2008;283(20):13565–13577.CrossRefGoogle ScholarPubMed
Yoshikawa, Y, Morita, M, Hosomi, H, et al. Knockdown of superoxide dismutase 2 enhances acetaminophen-induced hepatotoxicity in rat. Toxicology. 2009;264(1–2):89–95.CrossRefGoogle ScholarPubMed
Fujimoto, K, Kumagai, K, Ito, K, et al. Sensitivity of liver injury in heterozygous Sod2 knockout mice treated with troglitazone or acetaminophen. Toxicol Pathol. 2009;37(2):193–200.CrossRefGoogle ScholarPubMed
Alvarez-Sanchez, R, Montavon, F, Hartung, T, Pahler, A. Thiazolidinedione bioactivation: A comparison of the bioactivation potentials of troglitazone, rosiglitazone, and pioglitazone using stable isotope-labeled analogues and liquid chromatography tandem mass spectrometry. Chem Res Toxicol. 2006;19(8):1106–1116.CrossRefGoogle ScholarPubMed
Funk, C, Pantze, M, Jehle, L, et al. Troglitazone-induced intrahepatic cholestasis by an interference with the hepatobiliary export of bile acids in male and female rats. Correlation with the gender difference in troglitazone sulfate formation and the inhibition of the canalicular bile salt export pump (Bsep) by troglitazone and troglitazone sulfate. Toxicology. 2001;167(1):83–98.CrossRefGoogle ScholarPubMed
Lee, JK, Marion, T, Abe, K, Lim, C, Pollack, GM, Brouwer, KL. Hepatobiliary disposition of troglitazone and metabolites in rat and human sandwich-cultured hepatocytes: Use of Monte Carlo simulations to assess the impact of changes in biliary excretion on troglitazone sulfate accumulation. J Pharmacol Exp Ther. 2010:332(1):26–34.CrossRefGoogle ScholarPubMed
Uetrecht, J. Idiosyncratic drug reactions: Past, present, and future. Chem Res Toxicol. 2008;21(1):84–92.CrossRefGoogle ScholarPubMed
Evans, DC, Baillie, TA. Minimizing the potential for metabolic activation as an integral part of drug design. Curr Opin Drug Discov Devel. 2005;8(1):44–50.Google ScholarPubMed
Hanzlik, RP, Koen, YM, Theertham, B, Dong, Y, Fang, J. The reactive metabolite target protein database (TPDB) – A web-accessible resource. BMC Bioinformatics. 2007;8:95.CrossRefGoogle ScholarPubMed
Antoine, DJ, Williams, DP, Park, BK . Understanding the role of reactive metabolites in drug-induced hepatotoxicity: State of the science. Expert Opin Drug Metab Toxicol. 2008;4(11):1415–1427.CrossRefGoogle ScholarPubMed
Obach, RS, Kalgutkar, AS, Ryder, TF, Walker, GS. In vitro metabolism and covalent binding of enol-carboxamide derivatives and anti-inflammatory agents sudoxicam and meloxicam: Insights into the hepatotoxicity of sudoxicam. Chem Res Toxicol. 2008;21(9):1890–1899.CrossRefGoogle ScholarPubMed
Zhao, SX, Dalvie, DK, Kelly, JM, et al. NADPH-dependent covalent binding of [3H]paroxetine to human liver microsomes and S-9 fractions: Identification of an electrophilic quinone metabolite of paroxetine. Chem Res Toxicol. 2007;20(11): 1649–1657.CrossRefGoogle Scholar
Masubuchi, Y. Metabolic and non-metabolic factors determining troglitazone hepatotoxicity: A review. Drug Metab Pharmacokinet. 2006;21(5):347–356.CrossRefGoogle ScholarPubMed
Bauman, JN, Frederick, KS, Sawant, A, et al. Comparison of the bioactivation potential of the antidepressant and hepatotoxin nefazodone with aripiprazole, a structural analog and marketed drug. Drug Metab Dispos. 2008;36(6):1016–1029.CrossRefGoogle ScholarPubMed
Obach, RS, Kalgutkar, AS, Soglia, JR, Zhao, SX. Can in vitro metabolism-dependent covalent binding data in liver microsomes distinguish hepatotoxic from nonhepatotoxic drugs? An analysis of 18 drugs with consideration of intrinsic clearance and daily dose. Chem Res Toxicol. 2008;21(9):1814–1822.CrossRefGoogle ScholarPubMed
Bauman, JN, Kelly, JM, Tripathy, S, et al. Can in vitro metabolism-dependent covalent binding data distinguish hepatotoxic from nonhepatotoxic drugs? An analysis using human hepatocytes and liver S-9 fraction. Chem Res Toxicol. 2009;22(2):332–340.CrossRefGoogle ScholarPubMed
LeCluyse, EL, Alexandre, E, Hamilton, GA, et al. Isolation and culture of primary human hepatocytes. Method Mol Biol. 2005;290:207–229.Google ScholarPubMed
Hewitt, NJ, Lechon, MJ, Houston, JB, et al. Primary hepatocytes: Current understanding of the regulation of metabolic enzymes and transporter proteins, and pharmaceutical practice for the use of hepatocytes in metabolism, enzyme induction, transporter, clearance, and hepatotoxicity studies. Drug Metab Rev. 2007;39(1):159–234.CrossRefGoogle ScholarPubMed
Li, AP. Human hepatocytes as an effective alternative experimental system for the evaluation of human drug properties: General concepts and assay procedures. ALTEX. 2008;25(1):33–42.CrossRefGoogle ScholarPubMed
Li, N, Bi, YA, Duignan, DB, Lai, Y. Quantitative expression profile of hepatobiliary transporters in sandwich cultured rat and human hepatocytes. Mol Pharm. 2009;6(4):1180–1189.CrossRefGoogle ScholarPubMed
Xu, JJ, Diaz, D, O'Brien, PJ. Applications of cytotoxicity assays and pre-lethal mechanistic assays for assessment of human hepatotoxicity potential. Chem Biol Interact. 2004;150(1):115–128.CrossRefGoogle ScholarPubMed
Schmelzer, E, Acikgoez, A, Fruhauf, NR, et al. Biotransformation of cyclosporin in primary rat, porcine and human liver cell co-cultures. Xenobiotica. 2006;36(8):693–708.CrossRefGoogle ScholarPubMed
Dong, JQ, Smith, PC. Glucuronidation and covalent protein binding of benoxaprofen and flunoxaprofen in sandwich-cultured rat and human hepatocytes. Drug Metab Dispos. 2009;37(12):2314–2322.CrossRefGoogle ScholarPubMed
Kostrubsky, SE, Strom, SC, Kalgutkar, AS, et al. Inhibition of hepatobiliary transport as a predictive method for clinical hepatotoxicity of nefazodone. Toxicol Sci. 2006;90(2):451–459.CrossRefGoogle ScholarPubMed
Abe, K, Bridges, AS, Brouwer, KL. Use of sandwich-cultured human hepatocytes to predict biliary clearance of angiotensin II receptor blockers and HMG-CoA reductase inhibitors. Drug Metab Dispos. 2009;37(3):447–452.CrossRefGoogle ScholarPubMed
Smith, PJ, Blunt, N, Wiltshire, M, et al. Characteristics of a novel deep red/infrared fluorescent cell-permeant DNA probe, DRAQ5, in intact human cells analyzed by flow cytometry, confocal and multiphoton microscopy. Cytometry. 2000;40(4):280–291.3.0.CO;2-7>CrossRefGoogle ScholarPubMed
Donato, MT, Martinez-Romero, A, Jimenez, N, et al. Cytometric analysis for drug-induced steatosis in HepG2 cells. Chem Biol Interact. 2009;181(3):417–423.CrossRefGoogle ScholarPubMed
Stevenson, D, Wokosin, D, Girkin, J, Grant, MH. Measurement of the intracellular distribution of reduced glutathione in cultured rat hepatocytes using monochlorobimane and confocal laser scanning microscopy. Toxicol In Vitro. 2002;16(5):609–619.CrossRefGoogle ScholarPubMed
Dambach, DM, Andrews, BA, Moulin, F. New technologies and screening strategies for hepatotoxicity: Use of in vitro models. Toxicol Pathol. 2005;33(1):17–26.CrossRefGoogle ScholarPubMed
Gao, J, Ann Garulacan, L, Storm, SM, et al. Identification of in vitro protein biomarkers of idiosyncratic liver toxicity. Toxicol In Vitro. 2004;18(4):533–541.CrossRefGoogle ScholarPubMed
Xu, JJ, Henstock, PV, Dunn, MC, Smith, AR, Chabot, JR, Graaf, D. Cellular imaging predictions of clinical drug-induced liver injury. Toxicol Sci. 2008;105(1):97–105.CrossRefGoogle ScholarPubMed
Tukov, FF, Maddox, JF, Amacher, , Bobrowski, WF, Roth, RA, Ganey, PE. Modeling inflammation-drug interactions in vitro: A rat Kupffer cell-hepatocyte coculture system. Toxicol In Vitro. 2006;20(8):1488–1499.CrossRefGoogle ScholarPubMed
Khetani, SR, Bhatia, SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2008;26(1):120–126.CrossRefGoogle ScholarPubMed
Sivaraman, A, Leach, JK, Townsend, S, et al. A microscale in vitro physiological model of the liver: Predictive screens for drug metabolism and enzyme induction. Curr Drug Metab. 2005;6(6):569–591.CrossRefGoogle ScholarPubMed
Kidambi, S, Yarmush, RS, Novik, E, Chao, P, Yarmush, ML, Nahmias, Y. Oxygen-mediated enhancement of primary hepatocyte metabolism, functional polarization, gene expression, and drug clearance. Proc Natl Acad Sci USA. 2009;106(37):15714–15719.CrossRefGoogle ScholarPubMed

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
×