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.

  • Review Article
  • Published:

Pharmacogenetics and cancer therapy

Key Points

  • The field of pharmacogenetics has gained increased attention, particularly with the recent evidence that essentially every human gene is polymorphic. Polymorphisms in gene products that affect cancer drug metabolism can affect the toxicity or efficacy of anticancer therapy.

  • The 10% of the population harbouring mutant alleles of thiopurine methyltransferase are at increased risk of toxicity and secondary tumours after treatment with thiopurine drugs.

  • Polymorphisms in dihydropyrimidine dehydrogenase can affect the toxicity, and variations in thymidylate synthase can affect the anticancer efficacy, of the drug 5-fluorouracil.

  • A glucuronosyltransferase gene has a variable number of TA repeats in its promoter, which affect the expression levels of the enzyme. An active metabolite of a drug used to treat colon cancer, irinotecan, causes side effects more frequently in patients who have low glucuronosyltransferase on the basis of their promoter TA repeat status.

  • Glutathione transferases, enzymes that detoxify many cancer drugs, are inactivated in up to 50% of some populations.

  • Because oestrogen-replacement therapy in women is associated with a higher risk of endometrial cancer in patients who have a specific genotype in a steroid-metabolizing cytochrome P450, such pharmacogenetic information might assist in prescribing practices for oestrogen medications.

  • The data so far implicating genetic polymorphisms in cancer therapy support a practice of incorporating pharmacogenetic studies in cancer clinical trials.

Abstract

Pharmacogenetics is the study of how genetic variations affect drug response. These variations can affect a patient's response to cancer drugs, for which there is usually a fine line between a dosage that has a therapeutic effect and one that produces toxicity. Gaining better insight into the genetic elements of both the patient and the tumour that affect drug efficacy will eventually allow for individualized dosage determination and fewer adverse effects.

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

Access options

Buy this article

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

Figure 1: Relationship between cancer drug dose, antitumour efficacy and toxicity.
Figure 2: Types of polymorphisms that affect drug response.
Figure 3: Population distribution of drug-related polymorphisms.
Figure 4: Thiopurine methyltransferase polymorphisms that affect mercaptopurine therapy.
Figure 5: Polymorphisms that affect response to 5-fluorouracil (5-FU) therapy.
Figure 6: Polymorphisms that affect irinotecan therapy.
Figure 7: A single nucleotide polymorphism in intron 3 of CYP3A5 creates a nonfunctional enzyme.

Similar content being viewed by others

References

  1. Ratain, M. J. & Relling, M. V. Gazing into a crystal ball — cancer therapy in the post-genomic era. Nature Med. 7, 283–285 (2001).

    CAS  PubMed  Google Scholar 

  2. Klausner, R. D. Cancer, genomics, and the National Cancer Institute. J. Clin. Invest. 104, 15–17 (1999).

    Google Scholar 

  3. Vogel, F. Moderne probleme der humangenetik. Ergebnisse Inneren Medizin und Keinderheilkunde 12, 52–125 (1959).

    Google Scholar 

  4. Evans, W. E. & Relling, M. V. Pharmacogenomics: translating functional genomics into rational therapeutics. Science 286, 487–491 (1999).

    CAS  PubMed  Google Scholar 

  5. Spannbrucker, N., Eichelbaum, M., Steinke, B. and Dengler, H. J. A human genetic defect in the metabolism of sparteine. Verh. Dtsch. Ges. Inn. Med. 84, 1125–1127 (1978).

    Google Scholar 

  6. Gonzalez, F. J. et al. Characterization of the common genetic defect in humans deficient in debrisoquine metabolism. Nature 331, 442–446 (1988).The genetic basis underlying individual variations in responses to the antihypertensive drug debrisoquine. Poor drug metabolizers were found to express negligible amounts of the cytochrome P450 enzyme P450db1. The authors cloned the human P450db1 cDNA and purified the protein, discovering one of the most commonly occuring mutations. This was the start of modern pharmacogenetics.

    CAS  PubMed  Google Scholar 

  7. Mortimer, O. et al. Polymorphic formation of morphine from codeine in poor and extensive metabolizers of dextromethorphan: relationship to the presence of immunoidentified cytochrome P-450IID1. Clin. Pharmacol. Ther. 47, 27–35 (1990).

    CAS  PubMed  Google Scholar 

  8. Meyer, U. A. The genetic polymorphism of debrisoquine/sparteine metabolism-molecular mechanisms. Pharmacol. Ther. 46, 297–308 (1990).

    CAS  Google Scholar 

  9. Meyer, U. A. & Zanger, U. M. Molecular mechanisms of genetic polymorphisms of drug metabolism. Annu. Rev. Pharmacol. Toxicol. 37, 269–296 (1997).

    CAS  PubMed  Google Scholar 

  10. Johansson, I. et al. Inherited amplification of an active gene in the cytochrome P450 CYP2D locus as a cause of ultrarapid metabolism of debrisoquine. Proc. Natl Acad. Sci. USA 90, 11825–11829 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Sachidanandam, R. et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 409, 928–933 (2001).The work of several large collaborative groups, whose goal was to characterize the frequency and distribution characteristics of single nucleotide polymorphisms across the entire human genome.

    CAS  PubMed  Google Scholar 

  12. Pastinen, T. et al. A system for specific, high-throughput genotyping by allele-specific primer extension on microarrays. Genome Res. 10, 1031–1042 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Hoogendoorn, B. et al. Cheap, accurate and rapid allele frequency estimation of single nucleotide polymorphisms by primer extension and DHPLC in DNA pools. Hum. Genet. 107, 488–493 (2000).

    CAS  PubMed  Google Scholar 

  14. Giordano, M., Mellai, M., Hoogendoorn, B. & Momigliano-Richiardi, P. Determination of SNP allele frequencies in pooled DNAs by primer extension genotyping and denaturing high-performance liquid chromatography. J. Biochem. Biophys. Methods 47, 101–110 (2001).

    CAS  PubMed  Google Scholar 

  15. Mein, C. A. et al. Evaluation of single nucleotide polymorphism typing with invader on PCR amplicons and its automation. Genome Res. 10, 330–343 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Hessner, M. J., Budish, M. A. & Friedman, K. D. Genotyping of factor V G1691A (Leiden) without the use of PCR by invasive cleavage of oligonucleotide probes. Clin. Chem. 46, 1051–1056 (2000).

    CAS  PubMed  Google Scholar 

  17. Pfost, D. R., Boyce-Jacino, T. & Grant, D. M. A SNP shot: pharmacogenetics and the future of drug therapy. Trends Biotechnol. 18, 334–338 (2000).

    CAS  PubMed  Google Scholar 

  18. Pui, C. H. & Evans, W. E. Acute lymphoblastic leukemia. N. Engl. J. Med. 339, 605–615 (1998).

    CAS  PubMed  Google Scholar 

  19. Elion, G. B. The purine path to chemotherapy. Science 24, 441–447 (1989).

    Google Scholar 

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

    CAS  PubMed  Google Scholar 

  21. Lennard, L., Lilleyman, J. S., Van Loon, J. & Weinshilboum, R. M. Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemia. Lancet 336, 225–229 (1990).

    CAS  PubMed  Google Scholar 

  22. Lennard, L., Gibson, B. E., Nicole, T. & Lilleyman, J. S. Congenital thiopurine methyltransferase deficiency and 6-mercaptopurine toxicity during treatment for acute lymphoblastic leukaemia. Arch. Dis. Child 69, 577–579 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Dervieux, T. et al. Differing contribution of thiopurine methyltransferase to mercaptopurine versus thioguanine effects in human leukemic cells. Cancer Res. 61, 5810–5816 (2001).

    CAS  PubMed  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).This survey of 298 blood donors established a trimodal distribution for erythrocyte thiopurine methyltransferase activity, and classical family studies established that the three phenotypes were due to monogenic autosomal codominant inheritance. The authors suggested that the polymorphism could be used to predict response to thiopurine therapy, which was subsequently verified in many studies.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Tai, H. L. et al. Thiopurine S-methyltransferase deficiency: two nucleotide transitions define the most prevalent mutant allele associated with loss of catalytic activity in Caucasians. Am. J. Hum. Genet. 58, 694–702 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Otterness, D. et al. Human thiopurine methyltransferase pharmacogenetics: gene sequence polymorphisms. Clin. Pharmacol. Ther. 62, 60–73 (1997).

    CAS  PubMed  Google Scholar 

  27. 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).A molecular genotyping method showed over 95% concordance between thiopurine S –methyltransferase (TPMT) genotype and phenotype. The study was later extended to other populations with comparable success. TPMT genotyping eventually became the first certified molecular diagnostic for identifying patients requiring dosage adjustments of chemotherapy to avoid severe toxicity.

    CAS  PubMed  Google Scholar 

  28. Tai, H. L., Krynetski, E. Y., Schuetz, E. G., Yanishevski, Y. & Evans, W. E. Enhanced proteolysis of thiopurine S-methyltransferase (TPMT) encoded by mutant alleles in humans (TPMT*3A, TPMT*2): mechanisms for the genetic polymorphism of TPMT activity. Proc. Natl Acad. Sci. USA 94, 6444–6449 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Weinshilboum, R. M. Human pharmacogenetics: introduction. Fed. Proc. 43, 2295–2297 (1984).

    CAS  PubMed  Google Scholar 

  30. Evans, W. E. et al. Preponderance of thiopurine S-methyltransferase deficiency and heterozygosity among patients intolerant to mercaptopurine or azathioprine. J. Clin. Oncol. 19, 2293–2301 (2001).

    CAS  PubMed  Google Scholar 

  31. Dervieux, T. et al. Possible implication of thiopurine S–methyltransferase in the occurrence of infectious episodes during maintenance therapy of acute leukemia with mercaptopurine. Leukemia (in the press).

  32. Relling, M. V., Hancock, M. L., Boyett, J. M., Pui, C.-H. & Evans, W. E. Prognostic importance of 6-mercaptopurine dose intensity in acute lymphoblastic leukemia. Blood 93, 2817–2823 (1999).

    CAS  PubMed  Google Scholar 

  33. Relling, M. V. et al. High incidence of secondary brain tumours after radiotherapy and antimetabolites. Lancet 354, 34–39 (1999).Overall, the incidence of irradiation-induced brain tumours is usually low, occurring in only 1–2% of patients. However, almost half of the patients who received irradiation as well as mercaptopurine, and also possessed a germ-line defect in thiopurine methyltransferase, developed brain tumours. This shows how genetic characteristics and treatment interact to create risk groups.

    CAS  PubMed  Google Scholar 

  34. Relling, M. V. et al. Etoposide and antimetabolite pharmacology in patients who develop secondary acute myeloid leukemia. Leukemia 12, 346–352 (1998).

    CAS  PubMed  Google Scholar 

  35. Krynetskaia, N. F., Cai, X., Nitiss, J. L., Krynetski, E. Y. & Relling, M. V. Thioguanine substitution alters DNA cleavage mediated by topoisomerase II. FASEB J. 14, 2339–2344 (2000).

    CAS  PubMed  Google Scholar 

  36. Uribe-Luna, S. et al. Mutagenic consequences of the incorporation of 6-thioguanine into DNA. Biochem. Pharmacol. 54, 419–424 (1997).

    CAS  PubMed  Google Scholar 

  37. Pinedo, H. M. & Peters, G. F. Fluorouracil: biochemistry and pharmacology. J. Clin. Oncol. 6, 1653–1664 (1988).

    CAS  PubMed  Google Scholar 

  38. Heggie, G. D., Sommadossi, J. P., Cross, D. S., Huster, W. J. & Diasio, R. B. Clinical pharmacokinetics of 5-fluorouracil and its metabolites in plasma, urine, and bile. Cancer Res. 47, 2203–2206 (1987).

    CAS  PubMed  Google Scholar 

  39. Etienne, M. C. et al. Population study of dihydropyrimidine dehydrogenase in cancer patients. J. Clin. Oncol. 12, 2248–2253 (1994).

    CAS  PubMed  Google Scholar 

  40. Lu, Z., Zhang, R. & Diasio, R. B. Dihydropyrimidine dehydrogenase activity in human peripheral blood mononuclear cells and liver: population characteristics, newly identified deficient patients, and clinical implication in 5-fluorouracil chemotherapy. Cancer Res. 53, 5433–5438 (1993).

    CAS  PubMed  Google Scholar 

  41. Diasio, R. B., Beavers, T. L. & Carpenter, J. T. Familial deficiency of dihydropyrimidine dehydrogenase. Biochemical basis for familial pyrimidinemia and severe 5- fluorouracil-induced toxicity. J. Clin. Invest. 81, 47–51 (1988).The clinical observation of severe 5-fluorouracil toxicity in a patient served as the basis for determining its biochemical mechanism, a polymorphism that reduced dihydropyrimidine dehydrogenase activity. Family studies revealed the inherited basis for the defect.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Johnson, M. R. et al. Life-threatening toxicity in a dihydropyrimidine dehydrogenase-deficient patient after treatment with topical 5-fluorouracil. Clin. Cancer Res. 5, 2006–2011 (1999).

    CAS  PubMed  Google Scholar 

  43. Van Kuilenburg, A. B. et al. Lethal outcome of a patient with a complete dihydropyrimidine dehydrogenase (DPD) deficiency after administration of 5-fluorouracil: frequency of the common IVS14+1G→A mutation causing DPD deficiency. Clin. Cancer Res. 7, 1149–1153 (2001).

    CAS  PubMed  Google Scholar 

  44. Gonzalez, F. J. & Fernandez-Salguero, P. Diagnostic analysis, clinical importance and molecular basis of dihydropyrimidine dehydrogenase deficiency. Trends Pharmacol. Sci. 16, 325–327 (1995).

    CAS  PubMed  Google Scholar 

  45. Berger, R. et al. Dihydropyrimidine dehydrogenase deficiency leading to thymine-uraciluria. An inborn error of pyrimidine metabolism. Clin. Chim. Acta 141, 227–234 (1984).

    CAS  PubMed  Google Scholar 

  46. Wei, X., McLeod, H. L., McMurrough, J., Gonzalez, F. J. & Fernandez-Salguero, P. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5-fluorouracil toxicity. J. Clin. Invest. 98, 610–615 (1996).This was the first study to link a defect in dihyropyrimidine dehydrogenase (DPD) to 5-fluorouracil toxicity. A single point mutation at a splice site causes skipping of an exon, deletion of 55 amino acids and an inactive protein. Additional mutant alleles have now been linked to inheritance of DPD deficiency.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. McLeod, H. L. et al. Nomenclature for human DPYD alleles. Pharmacogenetics 8, 455–459 (1998).

    CAS  PubMed  Google Scholar 

  48. Hori, T. et al. Regional assignment of the human thymidylate synthase (TS) gene to chromosome band 18p11.32 by nonisotopic in situ hybridization. Hum. Genet. 85, 576–580 (1990).

    CAS  PubMed  Google Scholar 

  49. Villafranca, E. et al. Polymorphisms of the repeated sequences in the enhancer region of the thymidylate synthase gene promoter may predict downstaging after preoperative chemoradiation in rectal cancer. J. Clin. Oncol. 19, 1779–1786 (2001).This study shows that a variable number of tandem repeats in the thymidylate synthase promoter is associated with response to 5-fluorouracil. It provides evidence for further incorporation of pharmacogenetics studies in cancer clinical trials.

    CAS  PubMed  Google Scholar 

  50. Marsh, S., McKay, J. A., Cassidy, J. & McLeod, H. L. Polymorphism in the thymidylate synthase promoter enhancer region in colorectal cancer. Int. J. Oncol. 19, 383–386 (2001).

    CAS  PubMed  Google Scholar 

  51. Johnston, P. G. et al. Thymidylate synthase gene and protein expression correlate and are associated with response to 5-fluorouracil in human colorectal and gastric tumours. Cancer Res. 55, 1407–1412 (1995).

    CAS  PubMed  Google Scholar 

  52. Leichman, C. G. et al. Quantitation of intratumoural thymidylate synthase expression predicts for disseminated colorectal cancer response and resistance to protracted-infusion fluorouracil and weekly leucovorin. J. Clin. Oncol. 15, 3223–3229 (1997).

    CAS  PubMed  Google Scholar 

  53. Kawakami, K., Omura, K., Kanehira, E. & Watanabe, Y. Polymorphic tandem repeats in the thymidylate synthase gene is associated with its protein expression in human gastrointestinal cancers. Anticancer Res. 19, 3249–3252 (1999).

    CAS  PubMed  Google Scholar 

  54. Horie, N., Aiba, H., Oguro, K., Hojo, H. & Takeishi, K. Functional analysis and DNA polymorphism of the tandemly repeated sequences in the 5′-terminal regulatory region of the human gene for thymidylate synthase. Cell Struct. Funct. 20, 191–197 (1995).

    CAS  PubMed  Google Scholar 

  55. Rougier, P. et al. Randomised trial of irinotecan versus fluorouracil by continuous infusion after fluorouracil failure in patients with metastatic colorectal cancer. Lancet 352, 1407–1412 (1998).

    CAS  PubMed  Google Scholar 

  56. Kudoh, S. et al. Phase II study of irinotecan combined with cisplatin in patients with previously untreated small-cell lung cancer. West Japan Lung Cancer Group. J. Clin. Oncol. 16, 1068–1074 (1998).

    CAS  PubMed  Google Scholar 

  57. Gupta, E. et al. Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res. 54, 3723–3725 (1994).

    CAS  PubMed  Google Scholar 

  58. Kawato, Y., Aonuma, M., Hirota, Y., Kuga, H. & Sato, K. Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumour effect of CPT-11. Cancer Res. 51, 4187–4191 (1991).

    CAS  PubMed  Google Scholar 

  59. Bosma, P. J. et al. The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert's syndrome. N. Engl. J. Med. 333, 1171–1175 (1995).

    CAS  PubMed  Google Scholar 

  60. Ando, Y. et al. Polymorphisms of UDP-glucuronosyl-transferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res. 60, 6921–6926 (2000).

    CAS  PubMed  Google Scholar 

  61. Iyer, L. et al. Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J. Clin. Invest. 101, 847–854 (1998).The enzyme responsible for glucuronidation of the active metabolite of irinotecan was identified, and liver extracts taken from patients with an inherited deficiency in this enzyme were shown to be defective in this process. The Gunn rat model was used to confirm the role of the glucuronosyl-transferase.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Iyer, L. et al. Phenotype–genotype correlation of in vitro SN-38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism. Clin. Pharmacol. Ther. 65, 576–582 (1999).

    CAS  PubMed  Google Scholar 

  63. Fisher, M. B. et al. Tissue distribution and interindividual variation in human UDP-glucuronosyltransferase activity: relationship between UGT1A1 promoter genotype and variability in a liver bank. Pharmacogenetics 10, 727–739 (2000).

    CAS  PubMed  Google Scholar 

  64. MacKenzie, P. I. et al. The UDP glycosyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics 7, 255–269 (1997).

    CAS  PubMed  Google Scholar 

  65. Ritter, J. K. et al. A novel complex locus UGT1 encodes human bilirubin, phenol, and other UDP-glucuronosyltransferase isozymes with identical carboxyl termini. J. Biol. Chem. 267, 3257–3261 (1992).

    CAS  PubMed  Google Scholar 

  66. Wasserman, E. et al. Severe CPT-11 toxicity in patients with Gilbert's syndrome: two case reports. Ann. Oncol. 8, 1049–1051 (1997).

    CAS  PubMed  Google Scholar 

  67. Santos, A. et al. Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Clin. Cancer Res. 6, 2012–2020 (2000).

    CAS  PubMed  Google Scholar 

  68. Humerickhouse, R., Lohrbach, K., Li, L., Bosron, W. F. & Dolan, M. E. Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 and hCE-2. Cancer Res. 60, 1189–1192 (2000).

    CAS  PubMed  Google Scholar 

  69. Khanna, R., Morton, C. L., Danks, M. K. & Potter, P. M. Proficient metabolism of irinotecan by a human intestinal carboxylesterase. Cancer Res. 60, 4725–4728 (2000).

    CAS  PubMed  Google Scholar 

  70. Sugatani, J. et al. The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyl-transferase UGT1A1 gene and regulation by the nuclear receptor CAR. Hepatology 33, 1232–1238 (2001).

    CAS  PubMed  Google Scholar 

  71. Gupta, E., Wang, X., Ramirez, J. & Ratain, M. J. Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother. Pharmacol. 39, 440–444 (1997).

    CAS  PubMed  Google Scholar 

  72. Tew, K. D. Glutathione-associated enzymes in anticancer drug resistance. Cancer Res. 54, 4313–4320 (1994).

    CAS  PubMed  Google Scholar 

  73. Ketterer, B. Protective role of glutathione and glutathione transferases in mutagenesis and carcinogenesis. Mutat. Res. 202, 343–361 (1988).

    CAS  PubMed  Google Scholar 

  74. Nebert, D. W., McKinnon, R. A. & Puga, A. Human drug-metabolizing enzyme polymorphisms: effects on risk of toxicity and cancer. DNA Cell Biol. 15, 273–280 (1996).

    CAS  PubMed  Google Scholar 

  75. Woo, M. H. et al. Glutathione S-transferase genotypes in children who develop treatment-related acute myeloid malignancies. Leukemia 14, 226–231 (2000).

    Google Scholar 

  76. Chen, H. et al. Increased risk for myelodysplastic syndromes in individuals with glutathione transferase θ 1 (GSTT1) gene defect. Lancet 347, 295–297 (1996).

    CAS  PubMed  Google Scholar 

  77. Seidegard, J. & Ekstrom, G. The role of human glutathione transferases and epoxide hydrolases in the metabolism of xenobiotics. Environ. Health Perspect. 105, 791–799 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Chen, C.-L., Liu, Q. & Relling, M. V. Simultaneous characterization of glutathione S-transferase M1 and T1 polymorphisms by polymerase chain reaction in American whites and blacks. Pharmacogenetics 6, 187–191 (1996).

    CAS  PubMed  Google Scholar 

  79. Hayes, J. D. & Strange, R. C. Potential contribution of the glutathione S-transferase supergene family to resistance to oxidative stress. Free Radic. Res. 22, 193–207 (1995).

    CAS  PubMed  Google Scholar 

  80. Ban, N. et al. Transfection of glutathione S-transferase (GST)-π antisense complementary DNA increases the sensitivity of a colon cancer cell line to adriamycin, cisplatin, melphalan, and etoposide. Cancer Res. 56, 3577–3582 (1996).

    CAS  PubMed  Google Scholar 

  81. Stanulla, M., Schrappe, M., Brechlin, A. M., Zimmermann, M. & Welte, K. Polymorphisms within glutathione S-transferase genes (GSTM1, GSTT1, GSTP1) and risk of relapse in childhood B-cell precursor acute lymphoblastic leukemia: a case-control study. Blood 95, 1222–1228 (2000).

    CAS  PubMed  Google Scholar 

  82. Chen, C.-L. et al. Higher frequency of glutathione S-transferase deletions in black children with acute lymphoblastic leukemia. Blood 89, 1701–1707 (1997).

    CAS  PubMed  Google Scholar 

  83. Anderer, G. et al. Polymorphisms within glutathione S-transferase genes and initial response to glucocorticoids in childhood acute lymphoblastic leukaemia. Pharmacogenetics 10, 715–726 (2000).

    CAS  PubMed  Google Scholar 

  84. Sweeney, C. et al. Association between survival after treatment for breast cancer and glutathione S-transferase P1 Ile105Val polymorphism. Cancer Res. 60, 5621–5624 (2000).

    CAS  PubMed  Google Scholar 

  85. Davies, S. M. et al. Glutathione S-transferase polymorphisms and outcome of chemotherapy in childhood acute myeloid leukemia. J. Clin. Oncol. 19, 1279–1287 (2001).A randomized trial showed that intensive chemotherapy was more effective than conventionally administered chemotherapy. However, this study demonstrates that the benefit of the intensive schedule depends on polymorphisms in genes that encode glutathione transferase.

    PubMed  Google Scholar 

  86. Chabner, B. A., Donehower, C. & Schilsky, R. L. Clinical pharmacology of methotrexate. Cancer Treat. Rep. 65, 51–54 (1981).

    PubMed  Google Scholar 

  87. Frosst, P. et al. A candidate genetic risk factor for vascular disease: a common mutation in methylenetetrahydrofolate reductase. Nature Genet. 10, 111–113 (1995).A common point mutation affects the stability of methylene tetrahydrofolate reductase, an enzyme that regulates folate and homocysteine metabolism. Individuals homozygous for the mutation have significantly elevated plasma homocysteine levels and an increased risk of developing vascular disease. The mutation was subsequently shown to affect response to drugs such as methotrexate, and to increase the risk for congenital diseases such as spina bifida.

    CAS  PubMed  Google Scholar 

  88. Goyette, P. et al. Human methylenetetrahydrofolate reductase: isolation of cDNA, mapping and mutation identification. Nature Genet. 7, 195–200 (1994).

    CAS  PubMed  Google Scholar 

  89. Ulrich, C. M. et al. Pharmacogenetics of methotrexate: toxicity among marrow transplantation patients varies with the methylenetetrahydrofolate reductase C677T polymorphism. Blood 98, 231–234 (2001).

    CAS  PubMed  Google Scholar 

  90. Molloy, A. M. et al. Thermolabile variant of 5,10-methylenetetrahydrofolate reductase associated with low red-cell folates: implications for folate intake recommendations. Lancet 349, 1591–1593 (1997).

    CAS  PubMed  Google Scholar 

  91. Jacques, P. F., Selhub, J., Bostom, A. G., Wilson, P. W. & Rosenberg, I. H. The effect of folic acid fortification on plasma folate and total homocysteine concentrations. N. Engl. J. Med. 340, 1449–1454 (1999).

    CAS  PubMed  Google Scholar 

  92. Gorlick, R. et al. Intrinsic and acquired resistance to methotrexate in acute leukemia. N. Engl. J. Med. 335, 1041–1048 (1996).

    CAS  PubMed  Google Scholar 

  93. Jansen, G. et al. A structurally altered human reduced folate carrier with increased folic acid transport mediates a novel mechanism of antifolate resistance. J. Biol. Chem. 273, 30189–30198 (1998).

    CAS  PubMed  Google Scholar 

  94. Wong, S. C. et al. Impaired membrane transport in methotrexate-resistant CCRF-CEM cells involves early translation termination and increased turnover of a mutant reduced folate carrier. J. Biol. Chem. 274, 10388–10394 (1999).

    CAS  PubMed  Google Scholar 

  95. Chango, A. et al. A polymorphism (80G->A) in the reduced folate carrier gene and its associations with folate status and homocysteinemia. Mol. Genet. Metab. 70, 310–315 (2000).

    CAS  PubMed  Google Scholar 

  96. Carman, M. D. et al. Resistance to methotrexate due to gene amplification in a patient with acute leukemia. J. Clin. Oncol. 2, 16–20 (1984).

    CAS  PubMed  Google Scholar 

  97. Goto, Y. et al. A novel single-nucleotide polymorphism in the 3′-untranslated region of the human dihydrofolate reductase gene with enhanced expression. Clin. Cancer Res. 7, 1952–1956 (2001).

    CAS  PubMed  Google Scholar 

  98. Gonzalez, F. J. Evolution of the P450 gene superfamily: animal–plant 'warfare', molecular drive and human genetic differences in drug oxidation. Trends. Genet. 6, 182–186 (1990).

    CAS  PubMed  Google Scholar 

  99. Haiman, C. A., Hankinson, S. E., Colditz, G. A., Hunter, D. J. & De Vivo, I. A polymorphism in CYP17 and endometrial cancer risk. Cancer Res. 61, 3955–3960 (2001).

    CAS  PubMed  Google Scholar 

  100. McKean-Cowdin, R. et al. Risk of endometrial cancer and estrogen replacement therapy history by CYP17 genotype. Cancer Res. 61, 848–849 (2001).

    CAS  PubMed  Google Scholar 

  101. Chang, T. K., Yu, L., Maurel, P. & Waxman, D. J. Enhanced cyclophosphamide and ifosfamide activation in primary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxazaphosphorines. Cancer Res. 57, 1946–1954 (1997).

    CAS  PubMed  Google Scholar 

  102. Kivisto, K. T., Kroemer, H. K. & Eichelbaum, M. The role of human cytochrome P450 enzymes in the metabolism of anticancer agents: implications for drug interactions. Br. J. Clin. Pharmacol. 40, 523–530 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Relling, M. V. Are the major effects of P-glycoprotein modulators due to altered pharmacokinetics of anticancer drugs? Ther. Drug Monit. 18, 350–356 (1996).

    CAS  PubMed  Google Scholar 

  104. Kuehl, P. et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nature Genet. 27, 383–391 (2001).Although it had been known for many years that CYP3A5 is expressed in only 20–50% of human livers, this is the first elucidation of the molecular basis for variation in expression. The mechanism involves the common occurrance of a point mutation that results in alternative exon splicing and introduction of a stop codon.

    CAS  PubMed  Google Scholar 

  105. Paulussen, A. et al. Two linked mutations in transcriptional regulatory elements of the CYP3A5 gene constitute the major genetic determinant of polymorphic activity in humans. Pharmacogenetics 10, 415–424 (2000).

    CAS  PubMed  Google Scholar 

  106. Sata, F. et al. CYP3A4 allelic variants with amino acid substitutions in exon 7 and 12: Evidence for an allelic variant with altered catalytic activity. Clin. Pharmacol. Ther. 67, 48–56 (2000).

    CAS  PubMed  Google Scholar 

  107. Ball, S. E. et al. Population distribution and effects on drug metabolism of a genetic variant in the 5′ promoter region of CYP3A4. Clin. Pharmacol. Ther. 66, 288–294 (1999).

    CAS  PubMed  Google Scholar 

  108. Rebbeck, T. R., Jaffe, J. M., Walker, A. H., Wein, A. J. & Malkowicz, S. B. Modification of clinical presentation of prostate tumours by a novel genetic variant in CYP3A4. J. Natl Cancer Inst. 90, 1225–1229 (1998).

    CAS  PubMed  Google Scholar 

  109. Borst, P., Evers, R., Kool, M. & Wijnholds, J. A family of drug transporters: the multidrug resistance-associated proteins. J. Natl Cancer Inst. 92, 1295–1302 (2000).

    CAS  PubMed  Google Scholar 

  110. Norris, M. D. et al. Expression of the gene for multidrug-resistance-associated protein and outcome in patients with neuroblastoma. N. Engl. J. Med. 334, 231–238 (1996).

    CAS  PubMed  Google Scholar 

  111. Sullivan, G. F. et al. Regulation of expression of the multidrug resistance protein MRP1 by p53 in human prostate cancer cells. J. Clin. Invest. 105, 1261–1267 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Pirker, R. et al. MDR1 gene expression and treatment outcome in acute myeloid leukemia. J. Natl Cancer Inst. 83, 708–712 (1991).

    CAS  PubMed  Google Scholar 

  113. Ameyaw, M. M. et al. MDR1 pharmacogenetics: frequency of the C3435T mutation in exon 26 is significantly influenced by ethnicity. Pharmacogenetics 11, 217–221 (2001).

    CAS  PubMed  Google Scholar 

  114. Ito, S. et al. Polymorphism of the ABC transporter genes, MDR1, MRP1 and MRP2/cMOAT, in healthy Japanese subjects. Pharmacogenetics 11, 175–184 (2001).

    CAS  PubMed  Google Scholar 

  115. Hoffmeyer, S. et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc. Natl Acad. Sci. USA 97, 3473–3478 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Heuvel-Eibrink, M. M. et al. MDR1 gene-related clonal selection and P-glycoprotein function and expression in relapsed or refractory acute myeloid leukemia. Blood 97, 3605–3611 (2001).

    PubMed  Google Scholar 

  117. Carling, T., Rastad, J., Akerstrom, G. & Westin, G. Vitamin D receptor (VDR) and parathyroid hormone messenger ribonucleic acid levels correspond to polymorphic VDR alleles in human parathyroid tumours. J. Clin. Endocrinol. Metab. 83, 2255–2259 (1998).

    CAS  PubMed  Google Scholar 

  118. Ho, Y. V. et al. Polymorphism of the vitamin D receptor gene and corticosteroid-related osteoporosis. Osteoporos. Int. 9, 134–138 (1999).

    CAS  PubMed  Google Scholar 

  119. Huizenga, N. A. et al. A polymorphism in the glucocorticoid receptor gene may be associated with and increased sensitivity to glucocorticoids in vivo. J. Clin. Endocrinol. Metab 83, 144–151 (1998).

    CAS  PubMed  Google Scholar 

  120. Roden, D. M. Taking the 'idio' out of 'idiosyncratic': predicting torsades de pointes. Pacing Clin. Electrophysiol. 21, 1029–1034 (1998).

    CAS  PubMed  Google Scholar 

  121. Priori, S. G. et al. Genetic and molecular basis of cardiac arrhythmias: impact on clinical management part III. Circulation 99, 674–681 (1999).

    CAS  PubMed  Google Scholar 

  122. Rietschel, M. et al. Dopamine D3 receptor variant and tardive dyskinesia. Eur. Arch. Psychiatry Clin. Neurosci. 250, 31–35 (2000).

    CAS  PubMed  Google Scholar 

  123. Liggett, S. B. β2-Adrenergic receptor pharmacogenetics. Am. J. Respir. Crit. Care Med. 161, 197–201 (2000)

    Google Scholar 

  124. Arranz, M. J. et al. Pharmacogenetic prediction of clozapine response. Lancet 355, 1615–1616 (2000).Showed that up to 75% of the variation in the schizophrenia patients' response to the antipsychotic drug clozapine could be accounted for by just six genetic polymorphisms. This is a good example of how multiple polymorphisms in a relatively large population can be used to determine a complex drug response phenotype.

    CAS  PubMed  Google Scholar 

  125. O'Toole, L., Stewart, M., Padfield, P. & Channer, K. Effect of the insertion/deletion polymorphism of the angiotensin-converting enzyme gene on response to angiotensin-converting enzyme inhibitors in patients with heart failure. J. Cardiovasc. Pharm. 32, 988–994 (1998).

    CAS  Google Scholar 

  126. Martinelli, I. et al. High risk of cerebral-vein thrombosis in carriers of a prothrombin-gene mutation and in users of oral contraceptives. N. Engl. J. Med. 338, 1793–1797 (1998).

    CAS  PubMed  Google Scholar 

  127. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    CAS  PubMed  Google Scholar 

  128. Venter, J. C. et al. The sequence of the human genome. Science 291, 1304–1351 (2001).

    CAS  PubMed  Google Scholar 

  129. Kwok, P. Y. High-throughput genotyping assay approaches. Pharmacogenomics 1, 95–100 (2000).

    CAS  PubMed  Google Scholar 

  130. Klausner, R. D. The future of cancer research and the role of the National Cancer Institute. J. Clin. Oncol. 14, 2878–2883 (1996).

    CAS  PubMed  Google Scholar 

  131. Thomas, D. C. Design of gene characterization studies: an overview. J. Natl Cancer Inst. Monogr. 17–23 (1999).

  132. Spurdle, A. B. et al. CYP17 promoter polymorphism and breast cancer in Australian women under age forty years. J. Natl Cancer Inst. 92, 1674–1681 (2000).

    CAS  PubMed  Google Scholar 

  133. Mahgoub, A., Idle, J. R., Dring, L. G., Lancaster, R. & Smith, R. L. Polymorphic hydroxylation of debrisoquire in man. Lancet 2, 584–586 (1977).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported in part by the National Institutes of Health/National Institute of General Medical Sciences (NIH/NIGMS) Pharmacogenetics Research Network and Database, the National Cancer Institute (NCI), an NCI Core Grant, and the American Lebanese Syrian Associated Charities (ALSAC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mary V. Relling.

Supplementary information

Online tables

Online Table 1 | Common anticancer drugs and specific adverse effects (PDF 34 kb)

Online Table 2 | Examples of tumour genetic alterations associated with cancer drug resistance

Related links

Related links

DATABASES

CancerNet:

breast cancer

colorectal cancer

lymphoblastic leukaemia

myeloblastic leukaemias

 LocusLink:

CAR

carboxylesterase

CYP17

CYP2D6

CYP3A4

CYP3A

CYP3A5

DHFR

DPD

folylpolyglutamate synthase

γ-glutamyl hydrolase

glucocorticoid receptor

GST-A

GST-M1

GST-P1

GSTs

GST-T1

HPRT

MDR1

MRP1

MRP2

MTHFR

N-MYC

p53

SP1

topoisomerase II

TPMT

TS

UGT1A1

vitamin D receptor

 Medscape DrugInfo:

azathioprine

codeine

etoposide

5-fluorouracil

irinotecan

isoniazid

mercaptopurine

methotrexate

morphine

phenobarbital

prednisone

thioguanine

 OMIM:

Gilbert's syndrome

FURTHER INFORMATION

Cancer Genome Anatomy Project

Cancer trials

Human Genome Project Information

National Human Genome Institute

NCBI—SNP database

NIGMS site for the public on pharmacogenetics

Pharmacogenetics Knowledge Base

Pharmacogenetics of Anticancer Agents Research (PAAR) Group

SNP Consortium

Glossary

THERAPEUTIC RANGE

The range of doses associated with a reasonable probability of efficacy with an acceptable probability of toxicity.

ANTHRACYCLINES

A group of natural-product anticancer drugs that intercalate in DNA and inhibit topoisomerase II.

P-GLYCOPROTEIN

A cell membrane-embedded protein transporter, normally localized in epithelial tissues, the overexpression of which is linked to resistance of tumour cells to many natural-product anticancer drugs.

ETOPOSIDE-INDUCED SECONDARY LEUKAEMIAS

Drug-induced acute myeloid leukaemias that are characterized by a short onset time and balanced translocations of DNA.

HAEMOLYSIS

Lysis of red blood cells.

GLUCOSE-6-PHOSPHATE DEHYDROGENASE DEFICIENCY

An inherited enzyme deficiency associated with adverse drug effects that are precipitated by oxidative stress.

SUCCINYLCHOLINE

A neuromuscular blocking medication that induces paralysis.

GERM-LINE POLYMORPHISM

DNA sequence variation among individuals that is inherited and transmitted from one generation to the next.

CYTOCHROME P450

A member of a superfamily of haem-containing enzymes, most of which are concentrated in the endoplasmic reticulum of liver, where they oxidize small molecules.

PRIMER EXTENSION

A method to assess single nucleotide polymorphisms that is based on the annealing of an oligonucleotide primer immediately adjacent to the variable nucleotide, and addition of a complementary-base-specific labelled nucleotide ('sequencing one base at a time').

INVADER PROBE

A method to assess genetic polymorphisms that uses hybridization and 'invasion' of short DNA probes that hybridize to the target (variable versus wild-type) DNA. This causes the DNA to form a structure that can be recognized by a cleaving enzyme that releases a label for assay read-out.

MICROFLUIDICS

Technology that allows use of very small volumes of reagents, shortening reaction times and facilitating scale-up of molecular methods.

PRODRUG

A chemical with little or no intrinsic pharmacological activity that requires metabolism or biotransformation in order to exert is biological effects.

MYELOSUPPRESSION

Depressed production of blood cells deriving from the myeloid lineage, including platelets, some leukocytes and erythrocytes. Because many anticancer drugs suppress the growth or proliferation of rapidly dividing cells, myelosuppression is a common side effect.

NONSYNONYMOUS SNPS

Coding-region SNPs that change the amino-acid sequence of a protein.

TRANSITION

A type of nucleotide-pair substitution that involves the replacement of a purine with another purine, or of a pyrimidine with another pyrimidine: for example, G with A.

TOPOISOMERASE I

A nuclear protein that facilitates replication and transcription of DNA by cleaving single-strand DNA and thereby relaxing topological stress in supercoiled DNA.

LEUKOPAENIA

Low white blood cell (leukocyte) count.

CONSTITUTIVE ANDROSTANE RECEPTOR

A transcription factor that activates specific metabolic enzymes.

XENOBIOTIC

A foreign compound, not endogenously present in the body.

ELECTROPHILE

An electron-deficient atom or molecule that is drawn to react with electron-donating compounds.

PHARMACODYNAMICS

The relationship between drug concentration (pharmacokinetics) and its biological effects (what the drug does to the body).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Relling, M., Dervieux, T. Pharmacogenetics and cancer therapy. Nat Rev Cancer 1, 99–108 (2001). https://doi.org/10.1038/35101056

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/35101056

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