690
Views
0
CrossRef citations to date
0
Altmetric
Review

Genetic Characterization to Improve Interpretation and Clinical Management of Hepatotoxicity Caused by Tyrosine Kinase Inhibitors

, &
Pages 541-554 | Published online: 05 Apr 2013

References

  • Arora A , ScholarEM. Role of tyrosine kinase inhibitors in cancer therapy. J. Pharmacol. Exp. Ther.315 , 971–979 (2006).
  • Hartman JT , HapM, KoppHG et al. Tyrosine kinase inhibitors – a review on pharmacology, metabolism and side effects. Curr. Drug Metabol. 10 , 470–481 (2009).
  • Loriot Y , PerlemuterG, MalkaD et al. Drug insight: gastrointestinal and hepatic adverse events. Nat. Clin. Pract. Oncol. 5 , 268–278 (2008).
  • Chang J , RandM, BlumenthalG et al. FDA evaluation of hepatotoxicity related to tyrosine kinase inhibitors. J. Clin. Oncol. 29 , 3106 (2011).
  • Ostapowitz G , FontanaRJ, SchiotFD et al. Results of a prospective study of acute liver failure at 17 tertiary care centers in the United States. Ann. Intern. Med. 137 , 947–954 (2002).
  • Temple R . Hy‘s law: predicting serious hepatotoxicity. Pharmacoepidemiol. Drug Saf.15 , 241–243 (2006).
  • Lammert C , EinarssonS, SahaC et al. Relationship between daily dose of oral medications and idiosyncratic drug-induced liver injury: search for signals. Hepatology 47 , 2003–2009 (2008).
  • Howell BA , YangY, KumarR et al. In vitro to in vivo extrapolation and species response comparisons for drug-induced liver injury (DILI) using DILIsym™: a mechanistic, mathematical model of DILI. J. Pharmacokinet. Pharmacodyn.39 , 527–541 (2012).
  • Uetrecht J . Immune-mediated adverse drug reactions. Chem. Res. Toxicol.22 , 24–34 (2009).
  • Aithal GP , RawlinsMD, DayCP. Accuracy of hepatic adverse drug reaction reporting in one English health region. BMJ319 , 1541 (1999).
  • Bjornsson E . Drug-induced liver injury: Hy‘s rule revisited. Clin. Pharmacol. Ther.79 , 521–528 (2006).
  • Senior JR . Monitoring for hepatotoxicity: what is the predictive value of liver “function” tests? Clin. Pharmacol. Ther.85 , 331–334 (2009).
  • Bjornsson E , OlssonR. Outcome and prognostic markers in severe drug-induced liver disease. Hepatology42 , 481–489 (2005).
  • Andrade RJ , LucenaMI, FernandezMC et al. Drug-induced liver injury: an analysis of 461 incidences submitted to the Spanish registry over a 10-year period. Gastroenterology 129 , 512–521 (2005).
  • Zimmerman HJ . Hepatotoxicty: the Adverse Effects of Drugs and Other Chmicals on the Liver. Lippincott Williams & Wilkins, Philadelphia, PA, USA (1999).
  • Andrade RJ , LucenaMI, KaplowitzN et al. Outcome of acute idiosyncratic drug-induced liver injury: long term follow-up in a hepatotoxicity registry. Hepatology 44 , 1581–1588 (2006).
  • Aithal PG , DayCP. The natural history of histologically proved drug-induced liver disease. Gut44 , 731–735 (1999).
  • Sharma SK , SinglaR, SardaP et al. Safety of three dfferent reintroduction regimens of antituberculosis drugs after development of antituberculosis treatment-induced hepatotoxicity. Clin. Infect. Dis. 50 , 833–839 (2010).
  • Nordstrom BH , FraemanKH, LuoW et al. Liver function test abnormalities in patients treated with small molecule tyrosine kinase inhibitors. Presented at: International Society for Pharmacoepidemiology Annual Meeting, Brighton, UK, 19–22 August 2010.
  • O‘Brien SG , GuilhotF, LarsonRA et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N. Engl. J. Med. 348 , 995–1004 (2003).
  • Singer JB , ShouY, KantarjianHM et al. UGT1A1 promoter polymorphism increases risk of nilotinib-induced hyperbilirubinemia. Leukemia21 , 2311–2315 (2007).
  • Bonvin A , MesnilA, NicoliniFE et al. Dasatinib-induced acute hepatitis. Leuk. Lymphoma 49 , 1630–1632 (2008).
  • Carlini P , PapaldoP, FabiA et al. Liver toxicity after treatment with gefitinib and anastrazole: drug-drug interactions through cytochrome P450? J. Clin. Oncol. 35 , e60–e61 (2006).
  • Ho C , DavisJ, AndersonF et al. Side effects related to cancer treatment: CASE 1. Hepatitis hollowing treatment with gefitinib. J. Clin. Oncol. 23 , 8531–8533 (2005).
  • Shepherd FA , PereiraJR, CiuleanuT et al. Erlotinib in previously treated non-small cell lung cancer. N. Engl. J. Med. 353 , 123–132 (2005).
  • Arora A . Erlotinib-induced hepatotoxicity – clinical presentation and successful management: a case report. J. Clin. Exp. Hepatol.1 , 38–40 (2011).
  • Llovet JM , RicciS, MazzaferroV et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 359 , 378–390 (2008).
  • van Hootegem A , VerslypeC, van Steenbergen W. Sorafenib-induced liver failure: a case report and review of the literature. Case Rep. Hepatol.2011 , 1–4 (2011).
  • Mueller EW , RockeyMI, RashkinMC. Sunitinib-related fulminant hepatic failure: case report and review of the literature. Pharmacotherapy12 , 1066–1070 (2008).
  • Sternberg CA , DavisID, MardiakJ et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized Phase III trial. J. Clin. Oncol. 28 , 1061–1068 (2010).
  • Goodman V , WangQ, PanditeL et al. Incidence and management of hepatic toxicity in pazopanib-treated patients. Ann. Oncol. 21(Suppl. 8) , viii282 (2011).
  • Moy B , RappoldE, WilliamsL et al. Hepatobiliary abnormalities in patients with metastatic cancer treated with lapatinib. J. Clin. Oncol. 27 , 1043 (2009).
  • Kaplowitz N . Idiosyncratic drug hepatotoxicity. Nat. Rev. Drug Dis.4 , 489–499 (2005).
  • Tujios S , FontanaRJ. Mechanisms of drug-induced liver injury: from bedside to bench. Nat. Rev. Gastroenterol. Hepatol.8 , 202–211 (2011).
  • van Cutsem E , NowackiM, LangI et al. Randomized Phase III study of irinotecan and 5FU/FA with or without cetuximab in the first-line treatment of patients with metastatic colorectal cancer (mCRC): the CRYSTAL trial. J. Clin. Oncol. 25 , S164 (2007).
  • Srinivasan S , ParsaV, LiuCY et al. Trastuzumab-induced hepatotoxicity. Ann. Pharmacother. 42 , 1497–1501 (2008).
  • Munoz A , CarreraS, FerreiroJ et al. Reversible liver toxicity with adjuvant trastuzumab for localised breast cancer. Ann. Oncol. 18 , 2045–2046 (2007).
  • Pessaux P , PanaroF, CasnediC et al. Targeted molecular therapies (cetuximab and bevacizumab) do not induce additional hepatotoxicity: preliminary results of a case–control study. Eur. J. Surg. Oncol. 36 , 575–582 (2010).
  • Geyer CE , ForsterJ, LindquistD et al. Lapatinib pus capecitabine for HER2 positive advanced breast cancer. N. Engl. J. Med. 355 , 2733–2743 (2006).
  • Johnston S , PippenJ, PivotX et al. Lapatinib combined with letrozole compared with letrozole and placebo as first line therapy for postmenopausal horemone receptor-positive metastatic breast cancer. J. Clin. Oncol. 27 , 5538–5546 (2009).
  • Blackwell KL , BursteinHJ, StornioloAM et al. Randomized study of lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J. Clin. Oncol. 28 , 1124–1130 (2010).
  • Moy B , GossPE. Lapatinib-associated toxicity and practical management recommendations. Oncologist12 , 756–765 (2007).
  • Spraggs CF , BuddeLR, BrileyL et al. Hyperbilirubinemia in lapatinib treated patients is associated with Gilbert‘s syndrome UGT1A1 polymorphism. Cancer Res. 69(Suppl. 24) , Abstract 1112 (2009).
  • Landis S , ChenC, ByrneJA et al. Assessing physician compliance to recommended liver function test monitoring guidance in patients with metastatic breast cancer using lapatinib. Presented at: the 28th International Conference on Pharmacoepidemiology and Therapeutic Risk Management, CCIB Barcelona, Spain, 23–26 August 2012.
  • Polli JW , HumphreysJE, HarmonKA et al. The role of efflux and uptake transporters in N-{3-chloro-4-[(3-fluorobenzyl)oxy]phenyl}-6-[5-({[2-(methylsulfonyl)ethyl]amino}methyl)-2-furyl]-4-quinazolinamine GW572016, lapatinib) disposition and drug interactions. Drug Metab. Disp. 36 , 695–701 (2008).
  • Castellino S , O‘MaraM, KochK et al. Human metabolism of lapatinib, a dula kinase inhibitor: implications for hepatotoxicity. Drug Metab. Disp. 40 , 139–150 (2011).
  • Teng WC , OhJW, NewLS et al. Mechanism-based inactivation of cytochrome P450 3A4 by lapatinib. Mol. Pharmacol. 78 , 693–703 (2010).
  • Takakusa H , WahlinMD, ZhaoC et al. Metabolic-intermediate complex formation of human cytochrome P450 3A4 by lapatinib. Drug Metab. Disp. 39 , 1022–1030 (2011).
  • Chan EC , NewLS, ChuaTB et al. Interaction of lapatinib with cytochrome P450 3A5. Drug Metab. Disp. 40 , 1414–1422 (2012).
  • Pauli-Magnus C , MeierPJ. Hepatobiliary transporters and drug-induced cholestasis. Hepatology44 , 778–787 (2006).
  • Sonpavde G , HutsonTE. Pazopanib: a novel multitargeted tyrosine kinase inhibitor. Curr. Oncol. Rep.9 , 115–119 (2007).
  • Hutson TE , DavidID, MachielsJP et al. Efficacy and safety of pazopanib in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 28 , 475–480 (2010).
  • Xu CF , ReckBH, XueZ et al. Pazopanib-induced hyperbilirubinemia is associated with Gilbert‘s syndrome UGT1A1 polymorphism. Br. J. Cancer 102 , 1371–1377 (2010).
  • Goh BC , ReddyNJ, DandamudiUB et al. An evaluation of thr drug interaction potential of pazopanib, an oral vascular endothelial growth factor receptor tyeosine kinase inhibitor, using a modified cooperstown 5+1 cocktail in patients with advanced solid tumors. Clin. Pharmacol. Ther. 88 , 652–659 (2010).
  • Xu C , XueZ, BingN et al. Concomitant use of pazopanib and simvastatin increases the risk of transaminase elevations in patients with cancer. Ann. Oncol. 23 , 2470–2471 (2012).
  • Bosma PJ , ChowdhuryJR, BakkerC et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1A1 in Gilbert‘s syndrome. N. Engl. J. Med. 333 , 1171–1175 (1994).
  • Balisteri WF , JansenP, KarpenSJ et al. Intrahepatic cholestasis: summary of an American Association for the Study of Liver Diseases single-topic conference. Hepatology 42 , 222–235 (2005).
  • Sanna S , BusoneroF, MaschioA et al. Common variants in the SLCO1B3 locus are associated with bilirubin levels and unconjugated hyperbilirubinemia. Hum. Mol. Genet. 18 , 2711–2718 (2009).
  • Bielinski SJ , ChalHS, PathakJ et al. Mayo genome consortia: a genotype–phenotype resource for genome-wide association studies with an application to the analysis of circulating bilirubin levels. Mayo Clin. Proc. 86 , 606–614 (2011).
  • Kang TW , KimHJ, JuH et al. Genome-wide association of serum bilirubin levels in Korean population. Hum. Mol. Genet. 19(18) , 3672–3678 (2010).
  • Johnson AD , KavousiM, SmithAV et al. Genome-wide association meta-analysis for total serum bilirubin levels. Hum. Mol. Genet. 18 , 2700–2710 (2009).
  • Aithal GP , WatkinsPB, AndradeRJ et al. Case definition and phenotype standardisation in drug-induced liver injury. Clin. Pharmacol. Ther. 89 , 806–815 (2011).
  • Green RM , FlammS. AGA technical review on the evaluation of liver chemistry tests. Gastroenterology123 , 1367–1384 (2002).
  • Danoff TM , CampbellDA, McCarthyLC et al. A Gilbert‘s syndrome UGT1A1 variant confers susceptibility to tranilast-induced hyperbilirubinemia. Pharmacogenomics J. 4 , 49–53 (2004).
  • Lee JS , WangJ, MartinM et al. Genetic variation in UGT1A1 typical of Gilbert syndrome is associated with unconjugated hyperbilirubinemia in patients receiving tocilizumab. Pharmacogenet. Genomics 21 , 365–374 (2011).
  • Zucker SD , QinX, RousterSD et al. Mechanism of indinavir-induced hyperbilirubinemia. Proc. Natl Acad. Sci. USA 98 , 12671–12676 (2001).
  • Rotger M , TaffeP, BleiberG et al. Gilbert syndrome and the development of antiretroviral therapy–associated hyperbilirubinemia. J. Infect. Dis. 192 , 1381–1386 (2005).
  • Li L , BacanuS, WhittakerJC et al. Disentangling baseline and on-treatment total bilirubin associations with UGT1A1*28. Presented at: 60th Annual Meeting of the American Society for Human Genetics, Washington, DC, USA, 2–6 November 2010.
  • Daly AK , DayCP. Genetic association studies in drug-induced liver injury. Semin. Liver Dis.29 , 400–411 (2009).
  • Phillips EJ , MallalSA. Pharmacogenetics of drug hypersensitivity. Pharmacogenomics11 , 973–987 (2010).
  • Andrade RK , RoblesM, UlzrrunE et al. Drug-induced liver injury. Pharmacogenomics 10 , 1447–1487 (2009).
  • Daly AK , DonaldsonPT, BhatnagarP et al. HLA-B*5701 genotype is a major determinant of drug-induced liver injury due to flucloxacillin. Nat. Genet.41 , 816–819 (2009).
  • O‘Donohue J , OienKA, DonaldsonP et al. Co-amoxiclav jaundice: clinical and histological features and class II HLA association. Gut 47 , 717–720 (2000).
  • Lucena MI , MolokhiaM, ShenY et al. Susceptibility to amoxicillin–clavulanate-induced liver injury is influenced by multiple HLA class I and II alleles. Gastroenterology 141 , 338–347 (2011).
  • Hetherington S , HughesAR, MostellerM et al. Genetic variation in HLA-B region and hypersensitivity reactions to abacavir. Lancet 359 , 1121–1122 (2002).
  • Mallal SA , PhillipsEJ, CarosiG et al. HLA-B*5701 screening for hypersensitivity to abacavir. N. Engl. J. Med.358 , 568–579 (2008).
  • Chessman D , KostenkoL, LethborgT et al. Human leukocyte antigen class I-restricted activation of CD8+ T-cells provides the immunogenetic basis of a systemic drug hypersensitivity. Immunity 28 , 822–832 (2008).
  • Chung WH , HungSI, HongHS et al. Medical genetics: a marker for Stevens–Johnson syndrome. Nature 428 , 486 (2004).
  • Ko TM , ChungWH, LinCH et al. Shared and restricted T-cell receptor use is crucial for carbamazepine-induced Stevens–Johnson syndrome. J. All. Clin. Immunol. 128 , 1266–1276 (2011).
  • Xu CF , ReckBH, GoodmanVL et al. Association of the hemochromatosis gene with pazopanib-induced transaminase elevation in renal cell carcinoma. J. Hepatol. 54 , 1237–1243 (2011).
  • Watkins PB , DesaiM, BerkowitzSD et al. Evaluation of drug-induced serious heaptotoxicity (eDISH): application of this data organisation approach to Phase III clinical trials of rivaroxiban after total hip or knee replacement surgery. Drug Saf. 34 , 248–252 (2011).
  • Allen KJ , GurrinLC, ConstantineCC et al. Iron-overload-related disease in HFE hereditary hemochromatosis. N. Engl. J. Med. 358 , 221–230 (2008).
  • Forooghian F , DasB. Anti-angiogenic effects of ribonucleic acid interference targeting vascular endothelial growth factor and hypoxia-inducible factor-1α. Am. J.Opthalmol.144 , 761–768 (2007).
  • Danan G , BenichouC. Causality assessment of adverse reactions to drugs I: a novel method based on the conclusions of international consensus meetings: application to drug-indiced liver injuries. J. Clin. Epidemiol.11 , 272–276 (1993).
  • Andrade RJ , RoblesM, LucenaMI et al. Rechallenge in drug-induced liver injury: the attractive hazard. Expert Opin. Drug Saf. 8 , 709–714 (2009).
  • Spraggs CF , BuddeLR, BrileyLP et al. HLA-DQA1*02:01 is a major risk factor for lapatinib-induced hepatotoxicity in women with advanced breast cancer. J. Clin. Oncol.29 , 667–673 (2011).
  • Kindmark A , JawaidA, HabronCG et al. Genome-wide pharmacogenetic investigation of a hepatic adverse event without clinical signs of immunopathology suggests an underlying immune pathogenesis. Pharmacogenomics J. 8 , 186–195 (2008).
  • Strautnieks SS , BymeJA, PawlikowskaL et al. Severe bile salt export pump deficiency: 82 different ABCB11 mutations in 109 families. Gastroenterology 134 , 1203–1214 (2008).
  • Kroetz DL , Pauli-MagnusC, HodgesLM et al. Sequence diversity and haplotype structure in the human ABCB1 (MDR1 multidrug resistance transporter) gene. Pharmacogenomics 13 , 481–494 (2003).
  • Kobayashi D , IeiriI, HirotaT et al. Functional assessment of ABCG2 (BCRP) gene polymorphisms to protein expression in human placenta. Drug Metab. Dispos. 33 , 94–101 (2005).
  • Furihata T , SatohN, OhishiT et al. Functional analysis of a mutation in the SLCO1B1 gene (c.1628T>G) identified in a Japanese patient with pravastatin induced myopathy. Pharmacogenomics J. 10 , 185–193 (2009).
  • Jones EY , FuggerL, StromingerJL et al. MHC class II proteins and disease: a structural perspective. Persp. Nat. Rev. Immunol. 6 , 271–282 (2006).
  • Spraggs CF , ParhamLR, HuntCM et al. Lapatinib-induced liver injury characterized by class II HLA and Gilbert‘s syndrome genotypes. Clin. Pharmacol. Ther. 91 , 647–652 (2012).
  • Becquemont L . HLA: a pharmacogenomics success story. Pharmacogenomics11 , 277–281 (2010).
  • Bharadwaj M , IllingP, TheodosisA et al. Drug hypersensitivity and human leukocyte antigens of the major histocompatibility complex. Annu. Rev. Pharmacol. Toxicol. 52 , 401–431 (2012).
  • Goss PE , SmithI, O‘ShaughnessyJ et al. Adjuvant lapatinib for women with early-stage HER2-positive breast cancer: a randomised, controlled, Phase 3 trial. Lancet Oncol. 14 , 88–96 (2013).
  • Keam SJ . Dasatinib in chronic myeloid leukemia and Philadelphia chromosome positive acute lymphoblastic leukemia. BioDrugs22 , 59–69 (2008).
  • Carlini P , PapaldoP, FabiA et al. Liver toxicity after treatment with gefitinib and anastrazole: drug–drug interactions through cytochrome P450? J. Clin. Oncol. 35 , e60–e61 (2006).
  • Munster PN , BrittenCD, MitaM 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. 13 , 1238–1245 (2007).
  • Feng B , XuJJ, MirelesR et al. Role of hepatic transporters in the disposition and hepatotoxicity of a HER2 tyrosine kinase inhibitor CP-724,714. Science 108 , 492–500 (2009).
  • Escudier B , PortaC, BonoP et al. Patient preference between pazopanib and sunitinib: results of a randomised double-blind, crossover study in patients with metastatic renal cell carcinoma (PISCES study NCT 01064310). J. Clin. Oncol. 30 , 277S (2012).

▪ 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.