Skip to main content

Part of the book series: Cancer Drug Discovery and Development™ ((CDD&D))

  • 849 Accesses

Summary

A goal of pharmacogenetics is the design of individualized therapy based on a patient’s genomic sequence to maximize response and minimize toxicity. Current chemotherapy is associated with serious, at times life-threatening, toxicity. Identifying heritable genetic variants responsible for chemotherapeutic toxicities is challenging because of its multigenic nature and the difficulty of studying chemotherapeutic toxicity using traditional genetic screening strategies, such as unaffected families.

Cell-based models using lymphoblastoid cell lines from individuals comprising large pedigrees are a means to overcome these limitations. Several investigators within the NIH Pharmacogenetics Research Network have used cell-based models, genome-wide mapping strategies including linkage, and association studies to search for genes responsible for toxicities and for responses associated with chemotherapy. By comparing the variations in chemotherapy-induced cytotoxicity in cell lines from within and among families, the extent to which genetics contributes to human variation in susceptibility to such cytotoxicity can be evaluated.

Centre d’ Etude du Polymorphisme Humain (CEPH) pedigrees have genetic information in the form of SNPs and microsatellites in the public domain, which allows for linkage analysis to identify genetic regions harboring genes contributing to cytotoxicity. Another cell-based model utilizes International HapMap cell lines to identify genetic variants and gene expression associated with chemotherapeutic agent–induced cytotoxicity. With extensive genotypic information and gene expression levels available for these cell lines, associations can be made between genotype and cytotoxicity, and between genotype and gene expression; and correlations can be made between gene expression and cytotoxicity. These studies enable the determination of genetic variants contributing to sensitivity to cytotoxicity through the modulation of gene expression.

There are a number of advantages to the use of CEPH cell lines for evaluating toxicities associated with chemotherapy. Use of cell lines avoids giving toxic chemotherapy to healthy volunteers; cells can be grown under identical conditions; and extensive genotype data for CEPH cell lines are publicly available, allowing investigators to utilize classical mapping techniques (e.g., linkage analysis, association) to study the genetic influence of human variation in drug sensitivity.

Despite the advantages of this cell-based system, limitations include that LCLs represent only one specific tissue type from unaffected individuals, which may or may not be appropriate when assessing a phenotypic effect that occurs in a different tissue (e.g., peripheral neuropathy, diarrhea). Another limitation is that most cell lines do not express cytochrome P450 genes and thus the contribution of metabolism and pharmacokinetics on drug affect is not taken into account. In spite of these limitations, these cell lines provide an opportunity for initial identification of important genes and variants that can be validated in further studies. This comprehensive approach can be broadly applied to other phenotypes that are measurable in cell lines, and for the first time allows the application of familial genetic analysis in the search for genes important in chemotherapeutic response and toxicity.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Spitz MR, Wu X, Mills G. Integrative epidemiology: from risk assessment to outcome prediction. J Clin Oncol 2005;23:267–275.

    Article  PubMed  Google Scholar 

  2. Walgren RA, Meucci MA, McLeod HL. Pharmacogenomic discovery approaches: will the real genes please stand up? J Clin Oncol 2005;23:7342–7349.

    Article  CAS  PubMed  Google Scholar 

  3. Shukla SJ, Dolan ME. Use of CEPH and non-CEPH lymphoblast cell lines in pharmacogenetic studies. Pharmacogenomics 2005;6:303–310.

    Article  CAS  PubMed  Google Scholar 

  4. Meucci MA, Marsh S, Watters JW et al. CEPH individuals are representative of the European American population: implications for pharmacogenetics. Pharmacogenomics 2005;6:59–63.

    Article  CAS  PubMed  Google Scholar 

  5. Dausset J, Cann H, Cohen D et al. Centre d’Etude du Polymorphisme Humain (CEPH): collaborative genetic mapping of the human genome. Genomics 1990;6:575–577.

    Article  CAS  PubMed  Google Scholar 

  6. Sugimoto M, Tahara H, Ide T et al. Steps involved in immortalization and tumorigenesis in human B-lymphoblastoid cell lines transformed by Epstein–Barr virus. Cancer Res 2004;64:3361–3364.

    Article  CAS  PubMed  Google Scholar 

  7. Miller G, Lipman M. Release of infectious Epstein–Barr virus by transformed marmoset leukocytes. Proc Natl Acad Sci USA 1973;70:190–194.

    Article  CAS  PubMed  Google Scholar 

  8. Henle W, Diehl V, Kohn G et al. Herpes-type virus and chromosome marker in normal leukocytes after growth with irradiated Burkitt cells. Science 1967;157:1064–1065.

    Article  CAS  PubMed  Google Scholar 

  9. Gipps EM, Kidson C. Cellular radiosensitivity: expression of an MS susceptibility gene? Neurology 1984;34:808–811.

    CAS  PubMed  Google Scholar 

  10. Imray FP, Smith PJ, Relf W et al. Wilms’ tumour: association with cellular sensitivity to mitomycin C in patients and first-degree relatives. Lancet 1984;1:1148–1151.

    Article  CAS  PubMed  Google Scholar 

  11. Poot M, Gollahon KA, Rabinovitch PS. Werner syndrome lymphoblastoid cells are sensitive to camptothecin-induced apoptosis in S-phase. Hum Genet 1999;104:10–14.

    Article  CAS  PubMed  Google Scholar 

  12. Cloos J, Reid CB, van der Sterre ML et al. A comparison of bleomycin-induced damage in lymphocytes and primary oral fibroblasts and keratinocytes in 30 subjects. Mutagenesis 1999;14:87–93.

    Article  CAS  PubMed  Google Scholar 

  13. Jen KY, Cheung VG. Transcriptional response of lymphoblastoid cells to ionizing radiation. Genome Res 2003;13:2092–2100.

    Article  CAS  PubMed  Google Scholar 

  14. Schork NJ, Gardner JP, Zhang L et al. Genomic association/linkage of sodium lithium counter-transport in CEPH pedigrees. Hypertension 2002;40:619–628.

    Article  CAS  PubMed  Google Scholar 

  15. Morley M, Molony CM, Weber TM et al. Genetic analysis of genome-wide variation in human gene expression. Nature 2004;430:743–747.

    Article  CAS  PubMed  Google Scholar 

  16. Cheung VG, Conlin LK, Weber TM et al. Natural variation in human gene expression assessed in lymphoblastoid cells. Nat Genet 2003;33:422–425.

    Article  CAS  PubMed  Google Scholar 

  17. Watters JW, Kraja A, Meucci MA et al. Genome-wide discovery of loci influencing chemotherapy cytotoxicity. Proc Natl Acad Sci USA 2004;101:11809–11814.

    Article  CAS  PubMed  Google Scholar 

  18. Dolan ME, Newbold KG, Nagasubramanian R et al. Heritability and linkage analysis of sensitivity to cisplatin-induced cytotoxicity. Cancer Res 2004;64:4353–4356.

    Article  CAS  PubMed  Google Scholar 

  19. Huang RS, Kistner EO, Bleibel WK et al. Effect of population and gender on chemotherapeutic agent-induced cytotoxicity. Mol Cancer Ther 2007;6:31–36.

    Article  CAS  PubMed  Google Scholar 

  20. Cheung VG, Spielman RS, Ewens KG et al. Mapping determinants of human gene expression by regional and genome-wide association. Nature 2005;437:1365–1369.

    Article  CAS  PubMed  Google Scholar 

  21. Correa CR, Cheung VG. Genetic variation in radiation-induced expression phenotypes. Am J Hum Genet 2004;75:885–890.

    Article  CAS  PubMed  Google Scholar 

  22. The International HapMap Consortium. A haplotype map of the human genome. Nature 2005;437:1299–1320.

    Article  Google Scholar 

  23. Brem RB, Yvert G, Clinton R et al. Genetic dissection of transcriptional regulation in budding yeast. Science 2002;296:752–755.

    Article  CAS  PubMed  Google Scholar 

  24. Lo HS, Wang Z, Hu Y et al. Allelic variation in gene expression is common in the human genome. Genome Res 2003;13:1855–1862.

    Article  CAS  PubMed  Google Scholar 

  25. Cheung VG. Polymorphic landscape of the human genome. Eur J Hum Genet 2005;13:133–135.

    Article  CAS  PubMed  Google Scholar 

  26. Lewis CM. Genetic association studies: design, analysis and interpretation. Brief Bioinform 2002;3:146–153.

    Article  CAS  PubMed  Google Scholar 

  27. Cardon LR, Bell JI. Association study designs for complex diseases. Nat Rev Genet 2001;2:91–99.

    Article  CAS  PubMed  Google Scholar 

  28. Spielman RS, Ewens WJ. The TDT and other family-based tests for linkage disequilibrium and association. Am J Hum Genet 1996;59:983–989.

    CAS  PubMed  Google Scholar 

  29. Kimchi-Sarfaty C, Oh JM, Kim IW et al. A “silent” polymorphism in the MDR1 gene changes substrate specificity. Science 2007;315:525–528.

    Article  CAS  PubMed  Google Scholar 

  30. Burdick JT, Chen WM, Abecasis GR et al. In silico method for inferring genotypes in pedigrees. Nat Genet 2006;38:1002–1004.

    Article  CAS  PubMed  Google Scholar 

  31. Schadt EE, Monks SA, Drake TA et al. Genetics of gene expression surveyed in maize, mouse and man. Nature 2003;422:297–302.

    Article  CAS  PubMed  Google Scholar 

  32. Stranger BE, Forrest MS, Clark AG et al. Genome–wide associations of gene expression variation in humans. PLoS Genet 2005;1:e78.

    Article  PubMed  Google Scholar 

  33. Spielman RS, Bastone LA, Burdick JT et al. Common genetic variants account for differences in gene expression among ethnic groups. Nat Genet 2007;39:226–231.

    Article  CAS  PubMed  Google Scholar 

  34. Storey JD, Madeoy J, Strout JL et al. Gene-expression variation within and among human populations. Am J Hum Genet 2007;80:502–509.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are supported by the Pharmacogenetics of Anticancer Agents Research (PAAR) and the Comprehensive Research on Expressed Alleles in Therapeutic Evaluation (CREATE) groups within the NIH Pharmacogenetics Research Network (GM63340, GM61393).

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2008 Humana Press, a part of Springer Science+Business Media, LLC

About this chapter

Cite this chapter

Dolan, M.E., McLeod, H. (2008). Cell-Based Models to Identify Genetic Variants Contributing to Anticancer Drug Response. In: Innocenti, F. (eds) Genomics and Pharmacogenomics in Anticancer Drug Development and Clinical Response. Cancer Drug Discovery and Development™. Humana Press. https://doi.org/10.1007/978-1-60327-088-5_2

Download citation

Publish with us

Policies and ethics