Skip to main content
Top
Published in: Drug Safety 7/2013

01-07-2013 | Review Article

Hepatotoxicity of Tyrosine Kinase Inhibitors: Clinical and Regulatory Perspectives

Authors: Rashmi R. Shah, Joel Morganroth, Devron R. Shah

Published in: Drug Safety | Issue 7/2013

Login to get access

Abstract

The introduction of small-molecule tyrosine kinase inhibitors (TKIs) in clinical oncology has transformed the treatment of certain forms of cancers. As of 31 March 2013, 18 such agents have been approved by the US Food and Drug Administration (FDA), 15 of these also by the European Medicines Agency (EMA), and a large number of others are in development or under regulatory review. Unexpectedly, however, their use has been found to be associated with serious toxic effects on a number of vital organs including the liver. Drug-induced hepatotoxicity has resulted in withdrawal from the market of many widely used drugs and is a major public health issue that continues to concern all the stakeholders. This review focuses on hepatotoxic potential of TKIs. The majority of TKIs approved to date are reported to induce hepatic injury. Five of these (lapatinib, pazopanib, ponatinib, regorafenib and sunitinib) are sufficiently potent in this respect as to require a boxed label warning. Onset of TKI-induced hepatotoxicity is usually within the first 2 months of initiating treatment, but may be delayed, and is usually reversible. Fatality from TKI-induced hepatotoxicity is uncommon compared to hepatotoxic drugs in other classes but may lead to long-term consequences such as cirrhosis. Patients should be carefully monitored for TKI-induced hepatotoxicity, the management of which requires individually tailored reappraisal of the risk/benefit. The risk is usually manageable by dose adjustment or a switch to a suitable alternative TKI. Confirmation of TKI-induced hepatotoxicity can present challenges in the presence of hepatic metastasis and potential drug interactions. Its diagnosis in a patient with TKI-sensitive cancer requires great care if therapy with the TKI suspected to be causal is to be modified or interrupted as a result. Post-marketing experience with drugs such as imatinib, lapatinib and sorafenib suggests that the hepatotoxic safety of all the TKIs requires diligent surveillance.
Literature
1.
go back to reference Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353:172–87.PubMedCrossRef Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353:172–87.PubMedCrossRef
2.
go back to reference Chen MH, Kerkela R, Force T. Mechanisms of cardiomyopathy associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118:84–95.PubMedCrossRef Chen MH, Kerkela R, Force T. Mechanisms of cardiomyopathy associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118:84–95.PubMedCrossRef
3.
go back to reference Amir E, Seruga B, Martinez-Lopez J, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29:2543–9.PubMedCrossRef Amir E, Seruga B, Martinez-Lopez J, et al. Oncogenic targets, magnitude of benefit, and market pricing of antineoplastic drugs. J Clin Oncol. 2011;29:2543–9.PubMedCrossRef
4.
go back to reference Shah RR, Morganroth J, Shah DR. Cardiovascular safety of tyrosine kinase inhibitors: with a special focus on cardiac repolarization (QT interval). Drug Saf. 2013. doi:10.1007/s40264-013-0047-5. Shah RR, Morganroth J, Shah DR. Cardiovascular safety of tyrosine kinase inhibitors: with a special focus on cardiac repolarization (QT interval). Drug Saf. 2013. doi:10.​1007/​s40264-013-0047-5.
6.
go back to reference Seruga B, Sterling L, Wang L, et al. Reporting of serious adverse drug reactions of targeted anticancer agents in pivotal phase III clinical trials. J Clin Oncol. 2011;29:174–85.PubMedCrossRef Seruga B, Sterling L, Wang L, et al. Reporting of serious adverse drug reactions of targeted anticancer agents in pivotal phase III clinical trials. J Clin Oncol. 2011;29:174–85.PubMedCrossRef
7.
go back to reference Niraula S, Seruga B, Ocana A, et al. The price we pay for progress: a meta-analysis of harms of newly approved anticancer drugs. J Clin Oncol. 2012;30:3012–9.PubMedCrossRef Niraula S, Seruga B, Ocana A, et al. The price we pay for progress: a meta-analysis of harms of newly approved anticancer drugs. J Clin Oncol. 2012;30:3012–9.PubMedCrossRef
8.
go back to reference Björnsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology. 2005;42:481–9.PubMedCrossRef Björnsson E, Olsson R. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology. 2005;42:481–9.PubMedCrossRef
9.
go back to reference 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. Erratum in: Gastroenterology. 2005;129:1808. 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. Erratum in: Gastroenterology. 2005;129:1808.
10.
go back to reference Shah RR. Drug-induced hepatotoxicity: pharmacokinetic perspectives and strategies for risk reduction. Advers Drug React Toxicol Rev. 1999;18:181–233. Shah RR. Drug-induced hepatotoxicity: pharmacokinetic perspectives and strategies for risk reduction. Advers Drug React Toxicol Rev. 1999;18:181–233.
11.
go back to reference Srivastava A, Maggs JL, Antoine DJ, et al. Role of reactive metabolites in drug-induced hepatotoxicity. Handb Exp Pharmacol. 2010;196:165–94.PubMedCrossRef Srivastava A, Maggs JL, Antoine DJ, et al. Role of reactive metabolites in drug-induced hepatotoxicity. Handb Exp Pharmacol. 2010;196:165–94.PubMedCrossRef
12.
go back to reference Russmann S, Kullak-Ublick GA, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr Med Chem. 2009;16:3041–53.PubMedCrossRef Russmann S, Kullak-Ublick GA, Grattagliano I. Current concepts of mechanisms in drug-induced hepatotoxicity. Curr Med Chem. 2009;16:3041–53.PubMedCrossRef
13.
go back to reference Andrade RJ, Robles M, Ulzurrun E, et al. Drug-induced liver injury: insights from genetic studies. Pharmacogenomics. 2009;10:1467–87.PubMedCrossRef Andrade RJ, Robles M, Ulzurrun E, et al. Drug-induced liver injury: insights from genetic studies. Pharmacogenomics. 2009;10:1467–87.PubMedCrossRef
14.
go back to reference Bessone F. Non-steroidal anti-inflammatory drugs: what is the actual risk of liver damage? World J Gastroenterol. 2010;16:5651–61.PubMedCrossRef Bessone F. Non-steroidal anti-inflammatory drugs: what is the actual risk of liver damage? World J Gastroenterol. 2010;16:5651–61.PubMedCrossRef
15.
go back to reference Russmann S, Jetter A, Kullak-Ublick GA. Pharmacogenetics of drug-induced liver injury. Hepatology. 2010;52:748–61.PubMedCrossRef Russmann S, Jetter A, Kullak-Ublick GA. Pharmacogenetics of drug-induced liver injury. Hepatology. 2010;52:748–61.PubMedCrossRef
16.
go back to reference Tujios S, Fontana RJ. Mechanisms of drug-induced liver injury: from bedside to bench. Nat Rev Gastroenterol Hepatol. 2011;8:202–11.PubMedCrossRef Tujios S, Fontana RJ. Mechanisms of drug-induced liver injury: from bedside to bench. Nat Rev Gastroenterol Hepatol. 2011;8:202–11.PubMedCrossRef
17.
go back to reference Ju C, Reilly T. Role of immune reactions in drug-induced liver injury (DILI). Drug Metab Rev. 2012;44:107–15.PubMedCrossRef Ju C, Reilly T. Role of immune reactions in drug-induced liver injury (DILI). Drug Metab Rev. 2012;44:107–15.PubMedCrossRef
18.
go back to reference Pessayre D, Fromenty B, Berson A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev. 2012;44:34–87.PubMedCrossRef Pessayre D, Fromenty B, Berson A, et al. Central role of mitochondria in drug-induced liver injury. Drug Metab Rev. 2012;44:34–87.PubMedCrossRef
19.
go back to reference Spriet-Pourra C, Auriche M. Drug withdrawal from sale. Scrip reports. Richmond: PJB; 1994. Spriet-Pourra C, Auriche M. Drug withdrawal from sale. Scrip reports. Richmond: PJB; 1994.
20.
go back to reference Shah RR. Can pharmacogenetics help rescue drugs withdrawn from the market? Pharmacogenomics. 2006;7:889–908.PubMedCrossRef Shah RR. Can pharmacogenetics help rescue drugs withdrawn from the market? Pharmacogenomics. 2006;7:889–908.PubMedCrossRef
21.
go back to reference Chalasani N, Björnsson E. Risk factors for idiosyncratic drug-induced liver injury. Gastroenterology. 2010;138:2246–59.PubMedCrossRef Chalasani N, Björnsson E. Risk factors for idiosyncratic drug-induced liver injury. Gastroenterology. 2010;138:2246–59.PubMedCrossRef
22.
go back to reference 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;41:816–9.PubMedCrossRef 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;41:816–9.PubMedCrossRef
23.
go back to reference Zimmerman HJ, Lewis JH, Ishak KG, et al. Ticrynafen-associated hepatic injury: analysis of 340 cases. Hepatology. 1984;4:315–23.PubMedCrossRef Zimmerman HJ, Lewis JH, Ishak KG, et al. Ticrynafen-associated hepatic injury: analysis of 340 cases. Hepatology. 1984;4:315–23.PubMedCrossRef
24.
go back to reference Graham DJ, Drinkard CR, Shatin D, et al. Liver enzyme monitoring in patients treated with troglitazone. JAMA. 2001;286:831–3.PubMedCrossRef Graham DJ, Drinkard CR, Shatin D, et al. Liver enzyme monitoring in patients treated with troglitazone. JAMA. 2001;286:831–3.PubMedCrossRef
25.
go back to reference Graham DJ, Green L, Senior JR, et al. Troglitazone-induced liver failure: a case study. Am J Med. 2003;114:299–306.PubMedCrossRef Graham DJ, Green L, Senior JR, et al. Troglitazone-induced liver failure: a case study. Am J Med. 2003;114:299–306.PubMedCrossRef
31.
go back to reference Lewis JH. The adaptive response (drug tolerance) helps to prevent drug-induced liver injury. Gastroenterol Hepat (NY). 2012;8:333–6. Lewis JH. The adaptive response (drug tolerance) helps to prevent drug-induced liver injury. Gastroenterol Hepat (NY). 2012;8:333–6.
32.
go back to reference Eder JP, Shapiro GI, Appleman LJ, et al. A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin Cancer Res. 2010;16:3507–16.PubMedCrossRef Eder JP, Shapiro GI, Appleman LJ, et al. A phase I study of foretinib, a multi-targeted inhibitor of c-Met and vascular endothelial growth factor receptor 2. Clin Cancer Res. 2010;16:3507–16.PubMedCrossRef
33.
go back to reference 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;108:492–500.PubMedCrossRef 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;108:492–500.PubMedCrossRef
34.
go back to reference Munster PN, Britten CD, Mita M, et al. First study of the safety, tolerability, and pharmacokinetics of CP-724,714 in patients with advanced malignant solid HER2-expressing tumors. Clin Cancer Res. 2007;13:1238–45.PubMedCrossRef Munster PN, Britten CD, Mita M, et al. First study of the safety, tolerability, and pharmacokinetics of CP-724,714 in patients with advanced malignant solid HER2-expressing tumors. Clin Cancer Res. 2007;13:1238–45.PubMedCrossRef
35.
go back to reference Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: a meta-analysis. Cancer Treat Rev. 2013;39:199–206.PubMedCrossRef Teo YL, Ho HK, Chan A. Risk of tyrosine kinase inhibitors-induced hepatotoxicity in cancer patients: a meta-analysis. Cancer Treat Rev. 2013;39:199–206.PubMedCrossRef
37.
go back to reference Spataro V. Nilotinib in a patient with postnecrotic liver cirrhosis related to imatinib. J Clin Oncol. 2011;29:e50–2.PubMedCrossRef Spataro V. Nilotinib in a patient with postnecrotic liver cirrhosis related to imatinib. J Clin Oncol. 2011;29:e50–2.PubMedCrossRef
38.
go back to reference Ridruejo E, Cacchione R, Villamil AG, et al. Imatinib-induced fatal acute liver failure. World J Gastroenterol. 2007;13:6608–11.PubMed Ridruejo E, Cacchione R, Villamil AG, et al. Imatinib-induced fatal acute liver failure. World J Gastroenterol. 2007;13:6608–11.PubMed
39.
go back to reference Kong JH, Yoo SH, Lee KE, et al. Early imatinib-mesylate-induced hepatotoxicity in chronic myelogenous leukaemia. Acta Haematol. 2007;118:205–8.PubMedCrossRef Kong JH, Yoo SH, Lee KE, et al. Early imatinib-mesylate-induced hepatotoxicity in chronic myelogenous leukaemia. Acta Haematol. 2007;118:205–8.PubMedCrossRef
40.
go back to reference Pariente A, Etcharry F, Cales V, et al. Imatinib mesylate-induced acute hepatitis in a patient treated for gastrointestinal stromal tumour. Eur J Gastroenterol Hepatol. 2006;18:785–7.PubMedCrossRef Pariente A, Etcharry F, Cales V, et al. Imatinib mesylate-induced acute hepatitis in a patient treated for gastrointestinal stromal tumour. Eur J Gastroenterol Hepatol. 2006;18:785–7.PubMedCrossRef
41.
go back to reference Fuster F, Medina L, Vallansot R, et al. Imatinib-induced toxic hepatitis: description of two cases and review of the literature. Artic Span Gastroenterol Hepatol. 2007;30:525–30.CrossRef Fuster F, Medina L, Vallansot R, et al. Imatinib-induced toxic hepatitis: description of two cases and review of the literature. Artic Span Gastroenterol Hepatol. 2007;30:525–30.CrossRef
42.
go back to reference Tonyali O, Coskun U, Yildiz R, et al. Imatinib mesylate-induced acute liver failure in a patient with gastrointestinal stromal tumors. Med Oncol. 2010;27:768–73.PubMedCrossRef Tonyali O, Coskun U, Yildiz R, et al. Imatinib mesylate-induced acute liver failure in a patient with gastrointestinal stromal tumors. Med Oncol. 2010;27:768–73.PubMedCrossRef
43.
go back to reference Weise AM, Liu CY, Shields AF. Fatal liver failure in a patient on acetaminophen treated with sunitinib malate and levothyroxine. Ann Pharmacother. 2009;43:761–6.PubMedCrossRef Weise AM, Liu CY, Shields AF. Fatal liver failure in a patient on acetaminophen treated with sunitinib malate and levothyroxine. Ann Pharmacother. 2009;43:761–6.PubMedCrossRef
46.
go back to reference Takeda M, Okamoto I, Fukuoka M, et al. Successful treatment with erlotinib after gefitinib-related severe hepatotoxicity. J Clin Oncol. 2010;28:e273–4.PubMedCrossRef Takeda M, Okamoto I, Fukuoka M, et al. Successful treatment with erlotinib after gefitinib-related severe hepatotoxicity. J Clin Oncol. 2010;28:e273–4.PubMedCrossRef
47.
go back to reference Seki N, Uematsu K, Shibakuki R, et al. Promising new treatment schedule for gefitinib responders after severe hepatotoxicity with daily administration. J Clin Oncol. 2006;24:3213–4.PubMedCrossRef Seki N, Uematsu K, Shibakuki R, et al. Promising new treatment schedule for gefitinib responders after severe hepatotoxicity with daily administration. J Clin Oncol. 2006;24:3213–4.PubMedCrossRef
48.
go back to reference Kijima T, Shimizu T, Nonen S, et al. Safe and successful treatment with erlotinib after gefitinib-induced hepatotoxicity: difference in metabolism as a possible mechanism. J Clin Oncol. 2011;29:e588–90.PubMedCrossRef Kijima T, Shimizu T, Nonen S, et al. Safe and successful treatment with erlotinib after gefitinib-induced hepatotoxicity: difference in metabolism as a possible mechanism. J Clin Oncol. 2011;29:e588–90.PubMedCrossRef
49.
go back to reference Lim SY, Ando S, Nishiyama K, et al. Multiple myeloma emerging after prolonged gefitinib treatment for non-small cell lung carcinoma. Case Rep Oncol. 2011;4:198–203.PubMedCrossRef Lim SY, Ando S, Nishiyama K, et al. Multiple myeloma emerging after prolonged gefitinib treatment for non-small cell lung carcinoma. Case Rep Oncol. 2011;4:198–203.PubMedCrossRef
50.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
51.
go back to reference Suzumura T, Kimura T, Kudoh S, et al. Comparison of adverse events of erlotinib with those of gefitinib in patients with non-small cell lung cancer: a case-control study in a Japanese population. Osaka City Med J. 2012;58:25–34.PubMed Suzumura T, Kimura T, Kudoh S, et al. Comparison of adverse events of erlotinib with those of gefitinib in patients with non-small cell lung cancer: a case-control study in a Japanese population. Osaka City Med J. 2012;58:25–34.PubMed
52.
go back to reference Suzumura T, Kimura T, Kudoh S, et al. Reduced CYP2D6 function is associated with gefitinib-induced rash in patients with non-small cell lung cancer. BMC Cancer. 2012;12:568.PubMedCrossRef Suzumura T, Kimura T, Kudoh S, et al. Reduced CYP2D6 function is associated with gefitinib-induced rash in patients with non-small cell lung cancer. BMC Cancer. 2012;12:568.PubMedCrossRef
53.
go back to reference Li J, Karlsson MO, Brahmer J, et al. CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst. 2006;98:1714–23.PubMedCrossRef Li J, Karlsson MO, Brahmer J, et al. CYP3A phenotyping approach to predict systemic exposure to EGFR tyrosine kinase inhibitors. J Natl Cancer Inst. 2006;98:1714–23.PubMedCrossRef
54.
go back to reference Donahower B, McCullough SS, Kurten R, et al. Vascular endothelial growth factor and hepatocyte regeneration in acetaminophen toxicity. Am J Physiol Gastrointest Liver Physiol. 2006;291:G102–9.PubMedCrossRef Donahower B, McCullough SS, Kurten R, et al. Vascular endothelial growth factor and hepatocyte regeneration in acetaminophen toxicity. Am J Physiol Gastrointest Liver Physiol. 2006;291:G102–9.PubMedCrossRef
55.
go back to reference Wang T, Shankar K, Ronis MJ, et al. Mechanisms and outcomes of drug- and toxicant-induced liver toxicity in diabetes. Crit Rev Toxicol. 2007;37:413–59.PubMedCrossRef Wang T, Shankar K, Ronis MJ, et al. Mechanisms and outcomes of drug- and toxicant-induced liver toxicity in diabetes. Crit Rev Toxicol. 2007;37:413–59.PubMedCrossRef
56.
go back to reference Nakagawa H, Maeda S, Hikiba Y, et al. Deletion of apoptosis signal-regulating kinase 1 attenuates acetaminophen-induced liver injury by inhibiting c-Jun N-terminal kinase activation. Gastroenterology. 2008;135:1311–21.PubMedCrossRef Nakagawa H, Maeda S, Hikiba Y, et al. Deletion of apoptosis signal-regulating kinase 1 attenuates acetaminophen-induced liver injury by inhibiting c-Jun N-terminal kinase activation. Gastroenterology. 2008;135:1311–21.PubMedCrossRef
57.
go back to reference Revuelta-Cervantes J, Mayoral R, Miranda S, et al. Protein tyrosine phosphatase 1B (PTP1B) deficiency accelerates hepatic regeneration in mice. Am J Pathol. 2011;178:1591–604.PubMedCrossRef Revuelta-Cervantes J, Mayoral R, Miranda S, et al. Protein tyrosine phosphatase 1B (PTP1B) deficiency accelerates hepatic regeneration in mice. Am J Pathol. 2011;178:1591–604.PubMedCrossRef
58.
go back to reference Han D, Shinohara M, Ybanez MD, et al. Signal transduction pathways involved in drug-induced liver injury. Handb Exp Pharmacol. 2010;196:267–310.PubMedCrossRef Han D, Shinohara M, Ybanez MD, et al. Signal transduction pathways involved in drug-induced liver injury. Handb Exp Pharmacol. 2010;196:267–310.PubMedCrossRef
59.
go back to reference Gupta-Abramson V, Troxel AB, Nellore A, et al. Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol. 2008;26:4714–9.PubMedCrossRef Gupta-Abramson V, Troxel AB, Nellore A, et al. Phase II trial of sorafenib in advanced thyroid cancer. J Clin Oncol. 2008;26:4714–9.PubMedCrossRef
60.
go back to reference Llanos L, Bellot P, Zapater P, et al. Acute hepatitis in a patient with cirrhosis and hepatocellular carcinoma treated with sorafenib. Am J Gastroenterol. 2009;104:257–8.PubMedCrossRef Llanos L, Bellot P, Zapater P, et al. Acute hepatitis in a patient with cirrhosis and hepatocellular carcinoma treated with sorafenib. Am J Gastroenterol. 2009;104:257–8.PubMedCrossRef
61.
go back to reference Fairfax B, Pratap S, Roberts I, et al. Fatal case of sorafenib-associated idiosyncratic hepatotoxicity in the adjuvant treatment of a patient with renal cell carcinoma. BMC Cancer. 2012;12:590.PubMedCrossRef Fairfax B, Pratap S, Roberts I, et al. Fatal case of sorafenib-associated idiosyncratic hepatotoxicity in the adjuvant treatment of a patient with renal cell carcinoma. BMC Cancer. 2012;12:590.PubMedCrossRef
62.
go back to reference Herden U, Fischer L, Schafer H, et al. Sorafenib-induced severe acute hepatitis in a stable liver transplant recipient. Transplantation. 2010;90:98–9.PubMedCrossRef Herden U, Fischer L, Schafer H, et al. Sorafenib-induced severe acute hepatitis in a stable liver transplant recipient. Transplantation. 2010;90:98–9.PubMedCrossRef
63.
go back to reference Schramm C, Schuch G, Lohse AW. Sorafenib-induced liver failure. Am J Gastroenterol. 2008;103:2162–3.PubMedCrossRef Schramm C, Schuch G, Lohse AW. Sorafenib-induced liver failure. Am J Gastroenterol. 2008;103:2162–3.PubMedCrossRef
64.
go back to reference Marks AB, Gerard R, Fournier P, et al. Sorafenib-induced hepatic encephalopathy. Ann Pharmacother. 2009;43:2121.PubMedCrossRef Marks AB, Gerard R, Fournier P, et al. Sorafenib-induced hepatic encephalopathy. Ann Pharmacother. 2009;43:2121.PubMedCrossRef
65.
go back to reference Van Hootegem A, Verslype A, Van Steenbergen W. Sorafenib-induced liver failure: a case report and review of the literature. Case Rep Hepatol. 2011; Article ID 941395, 4 pp. Van Hootegem A, Verslype A, Van Steenbergen W. Sorafenib-induced liver failure: a case report and review of the literature. Case Rep Hepatol. 2011; Article ID 941395, 4 pp.
67.
go back to reference Xu C-F, Reck BH, Xue Z, et al. Pazopanib-induced hyperbilirubinemia is associated with Gilbert’s syndrome UGT1A1 polymorphism. Br J Cancer. 2010;102:1371–7.PubMedCrossRef Xu C-F, Reck BH, Xue Z, et al. Pazopanib-induced hyperbilirubinemia is associated with Gilbert’s syndrome UGT1A1 polymorphism. Br J Cancer. 2010;102:1371–7.PubMedCrossRef
68.
69.
go back to reference Mandery K, Glaeser H, Fromm MF. Interaction of innovative small molecule drugs used for cancer therapy with drug transporters. Br J Pharmacol. 2012;165:345–62.PubMedCrossRef Mandery K, Glaeser H, Fromm MF. Interaction of innovative small molecule drugs used for cancer therapy with drug transporters. Br J Pharmacol. 2012;165:345–62.PubMedCrossRef
70.
go back to reference Haouala A, Widmer N, Duchosal MA, et al. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood. 2011;117:e75–87.PubMedCrossRef Haouala A, Widmer N, Duchosal MA, et al. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood. 2011;117:e75–87.PubMedCrossRef
71.
go back to reference Court MH, Duan SX, von Moltke LL, et al. Interindividual variability in acetaminophen glucuronidation by human liver microsomes: identification of relevant acetaminophen UDP-glucuronosyltransferase isoforms. J Pharmacol Exp Ther. 2001;299:998–1006.PubMed Court MH, Duan SX, von Moltke LL, et al. Interindividual variability in acetaminophen glucuronidation by human liver microsomes: identification of relevant acetaminophen UDP-glucuronosyltransferase isoforms. J Pharmacol Exp Ther. 2001;299:998–1006.PubMed
72.
go back to reference Mutlib AE, Goosen TC, Bauman JN, et al. Kinetics of acetaminophen glucuronidation by UDP-glucuronosyltransferases 1A1, 1A6, 1A9 and 2B15. Potential implications in acetaminophen-induced hepatotoxicity. Chem Res Toxicol. 2006;19:701–9.PubMedCrossRef Mutlib AE, Goosen TC, Bauman JN, et al. Kinetics of acetaminophen glucuronidation by UDP-glucuronosyltransferases 1A1, 1A6, 1A9 and 2B15. Potential implications in acetaminophen-induced hepatotoxicity. Chem Res Toxicol. 2006;19:701–9.PubMedCrossRef
73.
go back to reference Prescott LF. Kinetics and metabolism of paracetamol and phenacetin. Br J Clin Pharmacol. 1980;10(Suppl 2):291S–8S.PubMedCrossRef Prescott LF. Kinetics and metabolism of paracetamol and phenacetin. Br J Clin Pharmacol. 1980;10(Suppl 2):291S–8S.PubMedCrossRef
74.
75.
go back to reference Kostrubsky SE, Sinclair JF, Strom SC, et al. Phenobarbital and phenytoin increased acetaminophen hepatotoxicity due to inhibition of UDP-glucuronosyltransferases in cultured human hepatocytes. Toxicol Sci. 2005;87:146–55.PubMedCrossRef Kostrubsky SE, Sinclair JF, Strom SC, et al. Phenobarbital and phenytoin increased acetaminophen hepatotoxicity due to inhibition of UDP-glucuronosyltransferases in cultured human hepatocytes. Toxicol Sci. 2005;87:146–55.PubMedCrossRef
76.
go back to reference Liu Y, Ramirez J, House L, et al. Comparison of the drug-drug interactions potential of erlotinib and gefitinib via inhibition of UDP-glucuronosyltransferases. Drug Metab Dispos. 2010;38:32–9.PubMedCrossRef Liu Y, Ramirez J, House L, et al. Comparison of the drug-drug interactions potential of erlotinib and gefitinib via inhibition of UDP-glucuronosyltransferases. Drug Metab Dispos. 2010;38:32–9.PubMedCrossRef
77.
go back to reference Liu Y, Ramírez J, Ratain MJ. Inhibition of paracetamol glucuronidation by tyrosine kinase inhibitors. Br J Clin Pharmacol. 2011;71:917–20.PubMedCrossRef Liu Y, Ramírez J, Ratain MJ. Inhibition of paracetamol glucuronidation by tyrosine kinase inhibitors. Br J Clin Pharmacol. 2011;71:917–20.PubMedCrossRef
78.
go back to reference Laine JE, Auriola S, Pasanen M, et al. Acetaminophen bioactivation by human cytochrome P450 enzymes and animal microsomes. Xenobiotica. 2009;39:11–21.PubMedCrossRef Laine JE, Auriola S, Pasanen M, et al. Acetaminophen bioactivation by human cytochrome P450 enzymes and animal microsomes. Xenobiotica. 2009;39:11–21.PubMedCrossRef
81.
go back to reference Nassar I, Pasupati T, Judson JP, et al. Histopathological study of the hepatic and renal toxicity associated with the co-administration of imatinib and acetaminophen in a preclinical mouse model. Malays J Pathol. 2010;32:1–11.PubMed Nassar I, Pasupati T, Judson JP, et al. Histopathological study of the hepatic and renal toxicity associated with the co-administration of imatinib and acetaminophen in a preclinical mouse model. Malays J Pathol. 2010;32:1–11.PubMed
82.
go back to reference Kim DW, Tan EY, Jin Y, et al. Effects of imatinib mesylate on the pharmacokinetics of paracetamol (acetaminophen) in Korean patients with chronic myelogenous leukaemia. Br J Clin Pharmacol. 2011;71:199–206.PubMedCrossRef Kim DW, Tan EY, Jin Y, et al. Effects of imatinib mesylate on the pharmacokinetics of paracetamol (acetaminophen) in Korean patients with chronic myelogenous leukaemia. Br J Clin Pharmacol. 2011;71:199–206.PubMedCrossRef
83.
go back to reference Goodman VL, Rock EP, Dagher R, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007;13:1367–73.PubMedCrossRef Goodman VL, Rock EP, Dagher R, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007;13:1367–73.PubMedCrossRef
84.
go back to reference Lim AYL, Segarra I, Chakravarthi S, et al. Histopathology and biochemistry analysis of the interaction between sunitinib and paracetamol in mice. BMC Pharmacol. 2010;10:14.PubMedCrossRef Lim AYL, Segarra I, Chakravarthi S, et al. Histopathology and biochemistry analysis of the interaction between sunitinib and paracetamol in mice. BMC Pharmacol. 2010;10:14.PubMedCrossRef
85.
go back to reference Ohashi Y, Suzuki K, Sakurai M, et al. Safety analysis of eight patients treated with erlotinib after severe gefitinib-induced liver injury. Gan To Kagaku Ryoho. 2010;37:1307–11. (Article in Japanese).PubMed Ohashi Y, Suzuki K, Sakurai M, et al. Safety analysis of eight patients treated with erlotinib after severe gefitinib-induced liver injury. Gan To Kagaku Ryoho. 2010;37:1307–11. (Article in Japanese).PubMed
86.
go back to reference Ku GY, Chopra A. Lopes Gde L Jr. Successful treatment of two lung cancer patients with erlotinib following gefitinib-induced hepatotoxicity. Lung Cancer. 2010;70:223–5.PubMedCrossRef Ku GY, Chopra A. Lopes Gde L Jr. Successful treatment of two lung cancer patients with erlotinib following gefitinib-induced hepatotoxicity. Lung Cancer. 2010;70:223–5.PubMedCrossRef
87.
go back to reference Nagano T, Kotani Y, Kobayashi K, et al. Successful erlotinib treatment for a patient with gefitinib-related hepatotoxicity and lung adenocarcinoma refractory to intermittently administered gefitinib. Case Rep Pulmonol. 2011;2011:812972.PubMed Nagano T, Kotani Y, Kobayashi K, et al. Successful erlotinib treatment for a patient with gefitinib-related hepatotoxicity and lung adenocarcinoma refractory to intermittently administered gefitinib. Case Rep Pulmonol. 2011;2011:812972.PubMed
88.
go back to reference Kitade H, Yamada T, Igarashi S, et al. Efficacy of low-dose erlotinib against gefitinib-induced hepatotoxicity in a patient with lung adenocarcinoma harboring EGFR mutations. Gan To Kagaku Ryoho. 2013;40:79–81. (Article in Japanese).PubMed Kitade H, Yamada T, Igarashi S, et al. Efficacy of low-dose erlotinib against gefitinib-induced hepatotoxicity in a patient with lung adenocarcinoma harboring EGFR mutations. Gan To Kagaku Ryoho. 2013;40:79–81. (Article in Japanese).PubMed
89.
go back to reference Takeda M, Okamoto I, Tsurutani J, et al. Clinical impact of switching to a second EGFR-TKI after a severe AE related to a first EGFR-TKI in EGFR-mutated NSCLC. Jpn J Clin Oncol. 2012;42:528–33.PubMedCrossRef Takeda M, Okamoto I, Tsurutani J, et al. Clinical impact of switching to a second EGFR-TKI after a severe AE related to a first EGFR-TKI in EGFR-mutated NSCLC. Jpn J Clin Oncol. 2012;42:528–33.PubMedCrossRef
90.
go back to reference Nakatomi K, Nakamura Y, Tetsuya I, et al. Treatment with gefitinib after erlotinib-induced liver injury: a case report. J Med Case Rep. 2011;5:593.PubMedCrossRef Nakatomi K, Nakamura Y, Tetsuya I, et al. Treatment with gefitinib after erlotinib-induced liver injury: a case report. J Med Case Rep. 2011;5:593.PubMedCrossRef
91.
go back to reference Kunimasa K, Yoshioka H, Iwasaku M, et al. Successful treatment of non-small cell lung cancer with gefitinib after severe erlotinib-related hepatotoxicity. Intern Med. 2012;51:431–4.PubMedCrossRef Kunimasa K, Yoshioka H, Iwasaku M, et al. Successful treatment of non-small cell lung cancer with gefitinib after severe erlotinib-related hepatotoxicity. Intern Med. 2012;51:431–4.PubMedCrossRef
92.
go back to reference Chang SC, Chang CY, Chen CY, et al. Successful erlotinib rechallenge after gefitinib-induced acute interstitial pneumonia. J Thorac Oncol. 2010;5:1105–6.PubMedCrossRef Chang SC, Chang CY, Chen CY, et al. Successful erlotinib rechallenge after gefitinib-induced acute interstitial pneumonia. J Thorac Oncol. 2010;5:1105–6.PubMedCrossRef
93.
go back to reference Fukui T, Otani S, Hataishi R, et al. Successful rechallenge with erlotinib in a patient with EGFR-mutant lung adenocarcinoma who developed gefitinib-related interstitial lung disease. Cancer Chemother Pharmacol. 2010;65:803–6.PubMedCrossRef Fukui T, Otani S, Hataishi R, et al. Successful rechallenge with erlotinib in a patient with EGFR-mutant lung adenocarcinoma who developed gefitinib-related interstitial lung disease. Cancer Chemother Pharmacol. 2010;65:803–6.PubMedCrossRef
94.
go back to reference Koma Y, Matsuoka H, Yoshimatsu H, et al. Successful treatment with erlotinib after gefitinib-induced interstitial lung disease: a case report and literature review. Int J Clin Pharmacol Ther. 2012;50:760–4.PubMed Koma Y, Matsuoka H, Yoshimatsu H, et al. Successful treatment with erlotinib after gefitinib-induced interstitial lung disease: a case report and literature review. Int J Clin Pharmacol Ther. 2012;50:760–4.PubMed
95.
go back to reference Tammaro KA, Baldwin PD, Lundberg AS. Interstitial lung disease following erlotinib (Tarceva) in a patient who previously tolerated gefitinib (Iressa). J Oncol Pharm Pract. 2005;11:127–30.PubMedCrossRef Tammaro KA, Baldwin PD, Lundberg AS. Interstitial lung disease following erlotinib (Tarceva) in a patient who previously tolerated gefitinib (Iressa). J Oncol Pharm Pract. 2005;11:127–30.PubMedCrossRef
96.
go back to reference Lin NU, Sarantopoulos S, Stone JR, et al. Fatal hepatic necrosis following imatinib mesylate therapy. Blood. 2003;102:3455–6.PubMedCrossRef Lin NU, Sarantopoulos S, Stone JR, et al. Fatal hepatic necrosis following imatinib mesylate therapy. Blood. 2003;102:3455–6.PubMedCrossRef
97.
go back to reference Kikuchi S, Muroi K, Takahashi S, et al. Severe hepatitis and complete molecular response caused by imatinib mesylate: possible association of its serum concentration with clinical outcomes. Leuk Lymphoma. 2004;45:2349–51.PubMedCrossRef Kikuchi S, Muroi K, Takahashi S, et al. Severe hepatitis and complete molecular response caused by imatinib mesylate: possible association of its serum concentration with clinical outcomes. Leuk Lymphoma. 2004;45:2349–51.PubMedCrossRef
98.
go back to reference Cross TJ, Bagot C, Portmann B, et al. Imatinib mesylate as a cause of acute liver failure. Am J Hematol. 2006;81:189–92.PubMedCrossRef Cross TJ, Bagot C, Portmann B, et al. Imatinib mesylate as a cause of acute liver failure. Am J Hematol. 2006;81:189–92.PubMedCrossRef
99.
go back to reference James C, Trouette H, Marit G, et al. Histological features of acute hepatitis after imatinib mesylate treatment. Leukemia. 2003;17:978–9.PubMedCrossRef James C, Trouette H, Marit G, et al. Histological features of acute hepatitis after imatinib mesylate treatment. Leukemia. 2003;17:978–9.PubMedCrossRef
100.
go back to reference Ohyashiki K, Kuriyama Y, Nakajima A, et al. Imatinib mesylate-induced hepatotoxicity in chronic myeloid leukemia demonstrated focal necrosis resembling acute viral hepatitis. Leukemia. 2002;16:2160–1.PubMedCrossRef Ohyashiki K, Kuriyama Y, Nakajima A, et al. Imatinib mesylate-induced hepatotoxicity in chronic myeloid leukemia demonstrated focal necrosis resembling acute viral hepatitis. Leukemia. 2002;16:2160–1.PubMedCrossRef
101.
go back to reference Saif MW. Erlotinib-induced acute hepatitis in a patient with pancreatic cancer. Clin Adv Hematol Oncol. 2008;6:191–9.PubMed Saif MW. Erlotinib-induced acute hepatitis in a patient with pancreatic cancer. Clin Adv Hematol Oncol. 2008;6:191–9.PubMed
102.
go back to reference Ho C, Davis J, Anderson F, et al. Side effects related to cancer treatment: hepatitis following treatment with gefitinib. J Clin Oncol. 2005;23:8531–3.PubMedCrossRef Ho C, Davis J, Anderson F, et al. Side effects related to cancer treatment: hepatitis following treatment with gefitinib. J Clin Oncol. 2005;23:8531–3.PubMedCrossRef
Metadata
Title
Hepatotoxicity of Tyrosine Kinase Inhibitors: Clinical and Regulatory Perspectives
Authors
Rashmi R. Shah
Joel Morganroth
Devron R. Shah
Publication date
01-07-2013
Publisher
Springer International Publishing AG
Published in
Drug Safety / Issue 7/2013
Print ISSN: 0114-5916
Electronic ISSN: 1179-1942
DOI
https://doi.org/10.1007/s40264-013-0048-4

Other articles of this Issue 7/2013

Drug Safety 7/2013 Go to the issue