Role of pharmacogenetics for the improvement of cancer treatment

  • Vita Dolžan
  • Petra Bohanec-Grabar
  • Cristina Rodriguez-Antona
Keywords: cancer treatment, genetic variability, pharmacogenetics

Abstract

Background: Cancer chemotherapy is associated with a great heterogeneity in patient response that makes the prediction of tumor response and/or drug toxicity very difficult. Many factors such as tumor biology, patient’s age, sex and organ function, are known to affect the therapeutic effects of drugs. Although these factors may be considered in decisions on the treatment regimens, the same regimen may result in undertreatment and insufficient therapeutic efficacy in some patients, while it can lead to overtreatment and increased toxicity in other patients. There is increasing evidence that genetic variability in drug metabolizing enzymes and/or drug targets influences drug response and may have a great impact on treatment outcome. This may be especially important for drugs with a narrow therapetic window and high level of toxicity, such as anticancer drugs. Many studies have shown that genetic variation of drug metabolizing enzymes such as cytochromes P450 (CYPs), UDP-glucuronosyltransferase (UGT), glutathione transferases (GSTs), thiopurine methyltransferase (TPMT) or dihydropyrimidine dehydrogenase (DPYD), drug transporters such as p-glycoprotein (MDR1) or reduced folate carrier (RFC1) and drug targets such as thymidylate syntahse (TYMS) or 5,10-methylenetetrahydrofolate reductase (MTHFR) influence not only the pharmacokinetics and/or pharmacodynamics of anticancer drugs but also the outcome of cancer treatment. In addition, somatic variations in the cancer cells can also have an impact on the anti-cancer drug efficacy, such as epidermal growth factor receptor 2 protein (HER2) overexpression and epidermal growth factor receptor (EGFR) mutations.

Conclusions: By studying the genetic variability of drug response, pharmacogenetics offers the possibility to identify patients that could benefit most from a particular treatment as well as those at increased risk for severe toxicity thus holding promises for a more individualized cancer treatment.

Downloads

Download data is not yet available.

References

Oyama T, Kagawa N, Kunugita N, Kitagawa K, Ogawa M, Yamaguchi T, et al. Expression of cytochrome P450 in tumor tissues and its association with cancer development. Front Biosci 2004; 9: 1967–76.
Rooseboom M, Commandeur JN, Vermeulen NP. Enzyme-catalyzed activation of anticancer prodrugs. Pharmacol Rev 2004; 56: 53–102.
Nebert DW, Dalton TP. The role of cytochrome P450 enzymes in endogenous signalling pathways and environmental carcinogenesis. Nat Rev Cancer 2006; 6: 947–60.
Roos PH, Bolt HM. Cytochrome P450 interactions in human cancers: new aspects considering CYP1B1. Expert Opin Drug Metab Toxicol 2005; 1: 187–202.
McFadyen MC, Melvin WT, Murray GI. Cytochrome P450 enzymes: novel options for cancer therapeutics. Mol Cancer Ther 2004; 3: 363–71.
Wang J, Pitarque M, Ingelman-Sundberg M. 3'-UTR polymorphism in the human CYP2A6 gene affects mRNA stability and enzyme expression. Biochem Biophys Res Commun 2006; 340: 491–7.
Yoshida R, Nakajima M, Watanabe Y, Kwon JT, Yokoi T. Genetic polymorphisms in human CYP2A6 gene causing impaired nicotine metabolism. Br J Clin Pharmacol 2002; 54: 511–7.
Nakajima M, Kwon JT, Tanaka N, Zenta T, Yamamoto Y, Yamamoto H, et al. Relationship between interindividual differences in nicotine metabolism and CYP2A6 genetic polymorphism in humans. Clin Pharmacol Ther 2001; 69: 72–8.
Oscarson M. Genetic polymorphisms in the cytochrome P450 2A6 (CYP2A6) gene: implications for interindividual differences in nicotine metabolism. Drug Metab Dispos 2001; 29: 91–5.
Gambier N, Batt AM, Marie B, Pfister M, Siest G, Visvikis-Siest S. Association of CYP2A6*1B genetic variant with the amount of smoking in French adults from the Stanislas cohort. Pharmacogenomics J 2005; 5: 271–5.
Oscarson M, McLellan RA, Gullsten H, Agundez JA, Benitez J, Rautio A, et al. Identification and characterisation of novel polymorphisms in the CYP2A locus: implications for nicotine metabolism. FEBS Lett 1999; 460: 321–7.
Malaiyandi V, Sellers EM, Tyndale RF. Implications of CYP2A6 genetic variation for smoking behaviors and nicotine dependence. Clin Pharmacol Ther 2005; 77: 145–58.
Ariyoshi N, Miyamoto M, Umetsu Y, Kunitoh H, Dosaka-Akita H, Sawamura Y, et al. Genetic polymorphism of CYP2A6 gene and tobacco-induced lung cancer risk in male smokers. Cancer Epidemiol Biomarkers Prev 2002; 11: 890–4.
Daigo S, Takahashi Y, Fujieda M, Ariyoshi N, Yamazaki H, Koizumi W, et al. A novel mutant allele of the CYP2A6 gene (CYP2A6*11 ) found in a cancer patient who showed poor metabolic phenotype towards tegafur. Pharmacogenetics 2002; 12: 299–306.
Komatsu T, Yamazaki H, Shimada N, Nakajima M, Yokoi T. Roles of cytochromes P450 1A2, 2A6, and 2C8 in 5-fluorouracil formation from tegafur, an anticancer prodrug, in human liver microsomes. Drug Metab Dispos 2000; 28: 1457–63.
Rae JM, Soukhova NV, Flockhart DA, Desta Z. Triethylenethiophosphoramide is a specific inhibitor of cytochrome P450 2B6: implications for cyclophosphamide metabolism. Drug Metab Dispos 2002; 30: 525–30.
Huang Z, Roy P, Waxman DJ. Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Biochem Pharmacol 2000; 59: 961–72.
Roy P, Yu LJ, Crespi CL, Waxman DJ. Development of a substrate-activity based approach to identify the major human liver P-450 catalysts of cyclophosphamide and ifosfamide activation based on cDNA-expressed activities and liver microsomal P-450 profiles. Drug Metab Dispos 1999; 27: 655–66.
Lang T, Klein K, Richter T, Zibat A, Kerb R, Eichelbaum M, et al. Multiple novel nonsynonymous CYP2B6 gene polymorphisms in Caucasians: demonstration of phenotypic null alleles. J Pharmacol Exp Ther 2004; 311: 34–43.
Gervot L, Rochat B, Gautier JC, Bohnenstengel F, Kroemer H, de Berardinis V, et al. Human CYP2B6: expression, inducibility and catalytic activities. Pharmacogenetics 1999; 9: 295–306.
Miksys S, Lerman C, Shields PG, Mash DC, Tyndale RF. Smoking, alcoholism and genetic polymorphisms alter CYP2B6 levels in human brain. Neuropharmacology 2003; 45: 122–32.
Yengi LG, Xiang Q, Pan J, Scatina J, Kao J, Ball SE, et al. Quantitation of cytochrome P450 mRNA levels in human skin. Anal Biochem 2003; 316: 103–10.
Wang H, Faucette S, Sueyoshi T, Moore R, Ferguson S, Negishi M, et al. A novel distal enhancer module regulated by pregnane X receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression. J Biol Chem 2003; 278: 14146–52.
Goodwin B, Moore LB, Stoltz CM, McKee DD, Kliewer SA. Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol 2001; 60: 427–31.
Xie HJ, Yasar U, Lundgren S, Griskevicius L, Terelius Y, Hassan M, et al. Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation. Pharmacogenomics J 2003; 3: 53–61.
Wang J, Sonnerborg A, Rane A, Josephson F, Lundgren S, Stahle L, et al. Identification of a novel specific CYP2B6 allele in Africans causing impaired metabolism of the HIV drug efavirenz. Pharmacogenet Genomics 2006; 16: 191–8.
Tsuchiya K, Gatanaga H, Tachikawa N, Teruya K, Kikuchi Y, Yoshino M, et al. Homozygous CYP2B6 *6 (Q172H and K262R) correlates with high plasma efavirenz concentrations in HIV-1 patients treated with standard efavirenz-containing regimens. Biochem Biophys Res Commun 2004; 319: 1322–6.
Hesse LM, He P, Krishnaswamy S, Hao Q, Hogan K, von Moltke LL, et al. Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by cytochrome P450 2B6 in human liver microsomes. Pharmacogenetics 2004; 14: 225–38.
Timm R, Kaiser R, Lotsch J, Heider U, Sezer O, Weisz K, et al. Association of cyclophosphamide pharmacokinetics to polymorphic cytochrome P450 2C19. Pharmacogenomics J 2005; 5: 365–73.
Takada K, Arefayene M, Desta Z, Yarboro CH, Boumpas DT, Balow JE, et al. Cytochrome P450 pharmacogenetics as a predictor of toxicity and clinical response to pulse cyclophosphamide in lupus nephritis. Arthritis Rheum 2004; 50: 2202–10.
Sim SC, Risinger C, Dahl ML, Aklillu E, Christensen M, Bertilsson L, et al. A common novel CYP2C19 gene variant causes ultrarapid drug metabolism relevant for the drug response to proton pump inhibitors and antidepressants. Clin Pharmacol Ther 2006; 79: 103–13.
Rahman A, Korzekwa KR, Grogan J, Gonzalez FJ, Harris JW. Selective biotransformation of taxol to 6 alpha-hydroxytaxol by human cytochrome P450 2C8. Cancer Res 1994; 54: 5543–6.
Kirchheiner J, Brockmoller J. Clinical consequences of cytochrome P450 2C9 polymorphisms. Clin Pharmacol Ther 2005; 77: 1–16.
Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical significance of the cytochrome P450 2C19 genetic polymorphism. Clin Pharmacokinet 2002; 41: 913–58.
Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J 2005; 5: 6–13.
Johansson I, Lundqvist E, Bertilsson L, Dahl ML, Sjoqvist F, Ingelman-Sundberg M. Inherited amplification of an active gene in the cytochrome P450 CYP2D locus as a cause of ultrarapid metabolism of debrisoquine. Proc Natl Acad Sci U S A 1993; 90: 11825–9.
Aklillu E, Persson I, Bertilsson L, Johansson I, Rodrigues F, Ingelman-Sundberg M. Frequent distribution of ultrarapid metabolizers of debrisoquine in an ethiopian population carrying duplicated and multiduplicated functional CYP2D6 alleles. J Pharmacol Exp Ther 1996; 278: 441–6.
Jin Y, Desta Z, Stearns V, Ward B, Ho H, Lee KH, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst 2005; 97: 30–9.
Goetz MP, Ingle JN, Couch FJ. Gene-expression-based predictors for breast cancer. N Engl J Med 2007; 356: 752; author reply – 3.
Kaiser R, Sezer O, Papies A, Bauer S, Schelenz C, Tremblay PB, et al. Patient-tailored antiemetic treatment with 5-hydroxytryptamine type 3 receptor antagonists according to cytochrome P450 2D6 genotypes. J Clin Oncol 2002; 20: 2805–11.
Engels FK, Ten Tije AJ, Baker SD, Lee CK, Loos WJ, Vulto AG, et al. Effect of cytochrome P450 3A4 inhibition on the pharmacokinetics of docetaxel. Clin Pharmacol Ther 2004; 75: 448–54.
Goh BC, Lee SC, Wang LZ, Fan L, Guo JY, Lamba J, et al. Explaining interindividual variability of docetaxel pharmacokinetics and pharmacodynamics in Asians through phenotyping and genotyping strategies. J Clin Oncol 2002; 20: 3683–90.
Hirth J, Watkins PB, Strawderman M, Schott A, Bruno R, Baker LH. The effect of an individual’s cytochrome CYP3A4 activity on docetaxel clearance. Clin Cancer Res 2000; 6: 1255–8.
Friedman HS, Petros WP, Friedman AH, Schaaf LJ, Kerby T, Lawyer J, et al. Irinotecan therapy in adults with recurrent or progressive malignant glioma. J Clin Oncol 1999; 17: 1516–25.
Published
2016-08-18
How to Cite
1.
Dolžan V, Bohanec-Grabar P, Rodriguez-Antona C. Role of pharmacogenetics for the improvement of cancer treatment. TEST ZdravVestn [Internet]. 18Aug.2016 [cited 5Aug.2024];76. Available from: http://vestnik-dev.szd.si/index.php/ZdravVest/article/view/1981
Section
Test Section

Most read articles by the same author(s)