Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy

A Corrigendum to this article was published on 26 April 2013

This article has been updated

Abstract

Cisplatin is a widely used and effective chemotherapeutic agent, although its use is restricted by the high incidence of irreversible ototoxicity associated with it1. In children, cisplatin ototoxicity is a serious and pervasive problem, affecting more than 60% of those receiving cisplatin2,3,4,5 and compromising language and cognitive development. Candidate gene studies have previously reported associations of cisplatin ototoxicity with genetic variants in the genes encoding glutathione S-transferases and megalin6,7,8. We report association analyses for 220 drug-metabolism genes in genetic susceptibility to cisplatin-induced hearing loss in children. We genotyped 1,949 SNPs in these candidate genes in an initial cohort of 54 children treated in pediatric oncology units, with replication in a second cohort of 112 children recruited through a national surveillance network for adverse drug reactions in Canada. We identified genetic variants in TPMT (rs12201199, P value = 0.00022, OR = 17.0, 95% CI 2.3–125.9) and COMT (rs9332377, P value = 0.00018, OR = 5.5, 95% CI 1.9–15.9) associated with cisplatin-induced hearing loss in children.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Cisplatin ototoxity and number of risk alleles.

Similar content being viewed by others

Change history

  • 21 March 2013

    In the version of this article initially published, the units for treatment duration in Table 1 were incorrectly given as weeks rather than months. This error has been corrected in the HTML and PDF versions of the article.

References

  1. Brock, P. & Bellman, S. Ototoxicity of cisplatinum. Br. J. Cancer 63, 159–160 (1991).

    Article  CAS  Google Scholar 

  2. Li, Y., Womer, R.B. & Silber, J.H. Predicting cisplatin ototoxicity in children: the influence of age and the cumulative dose. Eur. J. Cancer 40, 2445–2451 (2004).

    Article  CAS  Google Scholar 

  3. Coradini, P.P., Cigana, L., Selistre, S.G., Rosito, L.S. & Brunetto, A.L. Ototoxicity from cisplatin therapy in childhood cancer. J. Pediatr. Hematol. Oncol. 29, 355–360 (2007).

    Article  CAS  Google Scholar 

  4. Knight, K.R., Kraemer, D.F. & Neuwelt, E.A. Ototoxicity in children receiving platinum chemotherapy: underestimating a commonly occurring toxicity that may influence academic and social development. J. Clin. Oncol. 23, 8588–8596 (2005).

    Article  Google Scholar 

  5. Kushner, B.H., Budnick, A., Kramer, K., Modak, S. & Cheung, N.K. Ototoxicity from high-dose use of platinum compounds in patients with neuroblastoma. Cancer 107, 417–422 (2006).

    Article  CAS  Google Scholar 

  6. Oldenburg, J., Kraggerud, S.M., Cvancarova, M., Lothe, R.A. & Fossa, S.D. Cisplatin-induced long-term hearing impairment is associated with specific glutathione S-transferase genotypes in testicular cancer survivors. J. Clin. Oncol. 25, 708–714 (2007).

    Article  CAS  Google Scholar 

  7. Peters, U. et al. Glutathione S-transferase genetic polymorphisms and individual sensitivity to the ototoxic effect of cisplatin. Anti-Cancer Drugs. Anticancer Drugs 11, 639–643 (2000).

    Article  CAS  Google Scholar 

  8. Riedemann, L. et al. Megalin genetic polymorphisms and individual sensitivity to the ototoxic effect of cisplatin. Pharmacogenomics J. 8, 23–28 (2008).

    Article  CAS  Google Scholar 

  9. Blakley, B.W., Gupta, A.K., Myers, S.F. & Schwan, S. Risk factors for ototoxicity due to cisplatin. Arch. Otolaryngol. Head Neck Surg. 120, 541–546 (1994).

    Article  CAS  Google Scholar 

  10. Bess, F.H., Dodd-Murphy, J. & Parker, R.A. Children with minimal sensorineural hearing loss: prevalence, educational performance, and functional status. Ear Hear. 19, 339–354 (1998).

    Article  CAS  Google Scholar 

  11. Ekborn, A. et al. Cisplatin-induced hearing loss: influence of the mode of drug administration in the guinea pig. Hear. Res. 140, 38–44 (2000).

    Article  CAS  Google Scholar 

  12. Hirschhorn, J.N. & Daly, M.J. Genome-wide association studies for common diseases and complex traits. Nat. Rev. Genet. 6, 95–108 (2005).

    Article  CAS  Google Scholar 

  13. Ujiie, S., Sasaki, T., Mizugaki, M., Ishikawa, M. & Hiratsuka, M. Functional characterization of 23 allelic variants of thiopurine S-methyltransferase gene (TPMT*2 - *24). Pharmacogenet. Genomics 18, 887–893 (2008).

    Article  CAS  Google Scholar 

  14. Diatchenko, L. et al. Genetic basis for individual variations in pain perception and the development of a chronic pain condition. Hum. Mol. Genet. 14, 135–143 (2005).

    Article  CAS  Google Scholar 

  15. Nackley, A.G. et al. Human catechol-O-methyltransferase haplotypes modulate protein expression by altering mRNA secondary structure. Science 314, 1930–1933 (2006).

    Article  CAS  Google Scholar 

  16. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  Google Scholar 

  17. Visscher, H. et al. Application of principal component analysis to pharmacogenomic studies in Canada. Pharmacogenomics J. advance online publication, doi:10.1038/tpj.2009.36 (4 August 2009).

    Article  CAS  Google Scholar 

  18. The International HapMap Consortium. A haplotype map of the human genome. Nature 437, 1299–1320 (2005).

  19. Shord, S.S., Thompson, D.M., Krempl, G.A. & Hanigan, M.H. Effect of concurrent medications on cisplatin-induced nephrotoxicity in patients with head and neck cancer. Anticancer Drugs 17, 207–215 (2006).

    Article  CAS  Google Scholar 

  20. Krynetski, E.Y., Krynetskaia, N.F., Yanishevski, Y. & Evans, W.E. Methylation of mercaptopurine, thioguanine, and their nucleotide metabolites by heterologously expressed human thiopurine S-methyltransferase. Mol. Pharmacol. 47, 1141–1147 (1995).

    CAS  PubMed  Google Scholar 

  21. Yates, C.R. et al. Molecular diagnosis of thiopurine S-methyltransferase deficiency: genetic basis for azathioprine and mercaptopurine intolerance. Ann. Intern. Med. 126, 608–614 (1997).

    Article  CAS  Google Scholar 

  22. Relling, M.V. et al. Mercaptopurine therapy intolerance and heterozygosity at the thiopurine S-methyltransferase gene locus. J. Natl. Cancer Inst. 91, 2001–2008 (1999).

    Article  CAS  Google Scholar 

  23. Weinshilboum, R.M. Pharmacogenomics: catechol O-methyltransferase to thiopurine S-methyltransferase. Cell. Mol. Neurobiol. 26, 539–561 (2006).

    Article  CAS  Google Scholar 

  24. Weinshilboum, R.M. & Sladek, S.L. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am. J. Hum. Genet. 32, 651–662 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Ochoa, B., Bobadilla, N., Arrellin, G. & Herrera, L.A. S-Adenosyl-L-methionine increases serum BUN and creatinine in cisplatin-treated mice. Arch. Med. Res. 40, 54–58 (2009).

    Article  CAS  Google Scholar 

  26. Ahmed, Z.M. et al. Mutations of LRTOMT, a fusion gene with alternative reading frames, cause nonsyndromic deafness in humans. Nat. Genet. 40, 1335–1340 (2008).

    Article  CAS  Google Scholar 

  27. Du, X. et al. A catechol-O-methyltransferase that is essential for auditory function in mice and humans. Proc. Natl. Acad. Sci. USA 105, 14609–14614 (2008).

    Article  CAS  Google Scholar 

  28. Nelson, M.R. et al. Genome-wide approaches to identify pharmacogenetic contributions to adverse drug reactions. Pharmacogenomics J. 9, 23–33 (2008).

    Article  Google Scholar 

  29. Link, E. et al. SLCO1B1 variants and statin-induced myopathy—a genomewide study. N. Engl. J. Med. 359, 789–799 (2008).

    Article  CAS  Google Scholar 

  30. Ross, C.J. et al. Genotypic approaches to therapy in children: a national active surveillance network (GATC) to study the pharmacogenomics of severe adverse drug reactions in children. Ann. NY Acad. Sci. 1110, 177–192 (2007).

    Article  CAS  Google Scholar 

  31. Carlson, C.S. et al. Selecting a maximally informative set of single-nucleotide polymorphisms for association analyses using linkage disequilibrium. Am. J. Hum. Genet. 74, 106–120 (2004).

    Article  CAS  Google Scholar 

  32. Guo, S.W. & Thompson, E.A. Performing the exact test of Hardy-Weinberg proportion for multiple alleles. Biometrics 48, 361–372 (1992).

    Article  CAS  Google Scholar 

  33. Gao, X., Starmer, J. & Martin, E.R. A multiple testing correction method for genetic association studies using correlated single nucleotide polymorphisms. Genet. Epidemiol. 32, 361–369 (2008).

    Article  Google Scholar 

Download references

Acknowledgements

We especially want to acknowledge the study participants and their families for their participation in the Canadian Pharmacogenomics Network for Drug Safety (CPNDS) network; this work could not have been done without your help and support. We also want to acknowledge the support of the CPNDS active ADR surveillance consortium, as well as S. Langlois and B. Casey (Supplementary Note). The study was funded by Genome Canada, and additional funding was also provided by Genome British Columbia, Child and Family Research Institute, University of British Columbia Faculty of Pharmaceutical Sciences, Canadian Institutes of Health Research, Canada Foundation for Innovation, Canada Gene Cure Foundation, Canadian Society of Clinical Pharmacology, BC Clinical Genomics Network, C17 Research Network and Childhood Cancer Foundation–Candlelighters Canada, Michael Smith Foundation for Health Research, Health Canada, Pfizer, Eli Lilly, Merck Frosst and Janssen-Ortho. This work was funded as part of the peer-reviewed Genome Canada Applied Health Research Program; the pharmaceutical industry partners had no formal or informal role in this program of research.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

B.C.C., S.R.R., M.J.R., P.C.R. and members of the CPNDS consortium recruited subject cohorts. B.B., B.C.C. and S.R.R. phenotyped subject cohorts. C.J.D.R., H.K.-E., H.V., A.M.K.B. and M.S.P. designed and performed genotyping studies. C.J.D.R., H.K.-E., H.V., A.B., Y.F.-Z. and M.-P.D. analyzed genotyping data. B.C.C. and M.R.H. planned and coordinated the study. All authors contributed to the final version of the manuscript.

Corresponding author

Correspondence to Michael R Hayden.

Additional information

A full list of members is provided in the Supplementary Note.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Figures 1 and 2 and Supplementary Tables 2–5. (PDF 782 kb)

Supplementary Table 1

Allele frequencies and results of the case-control tests of association (XLS 803 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ross, C., Katzov-Eckert, H., Dubé, MP. et al. Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet 41, 1345–1349 (2009). https://doi.org/10.1038/ng.478

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.478

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing