Skip to main content
Top
Published in: Familial Cancer 3/2008

01-09-2008

Influence of germline polymorphisms of GSTT1, GSTM1, and GSTP1 in familial versus sporadic breast cancer susceptibility and survival

Authors: Volga S. Syamala, Leelakumari Sreeja, Vani Syamala, Praveenkumar B. Raveendran, Rajan Balakrishnan, Ratheesan Kuttan, Ravindran Ankathil

Published in: Familial Cancer | Issue 3/2008

Login to get access

Abstract

Identifying genes associated with familial inheritance of breast cancer continues to be a major goal of current research as the known high penetrance genes could be attributable for only a small percentage of the risk. So, it is hypothesized that the low penetrance genes may also modify the risk for familial breast cancer. In the present case–control study, undertaken to examine the influence of polymorphisms of GSTs in familial and sporadic breast cancer susceptibility, 597 women including 222 sporadic breast cancer patients, 125 familial breast cancer patients and 250 females with no history of cancer as controls were genotyped by PCR based methods. Odds Ratios (ORs) and 95% Confidence Intervals (95%CIs) were calculated by unconditional logistic regression adjusted to age. Interestingly, GSTM1 deletion was found to be significantly associated only with familial breast cancer (OR = 2.0; 95%CI = 1.252–3.128) while GSTT1 was associated only with sporadic breast cancer (OR = 2.3; 95%CI = 1.336–3.970). GSTP1 Ile105Val polymorphism was associated neither with sporadic nor familial breast cancer susceptibility (P value > 0.05). The GST genotypes did not have any effect on the survival of both familial and sporadic breast cancer patients. However, familial breast cancer patients with GSTM1 null genotype had a relative risk of 0.42 (95%CI = 0.18–0.97) for an advanced disease stage. The results indicate that, in addition to the known high penetrance genes, certain low penetrance genes may also play a role, in the familial inheritance of breast cancer. It is also noticed that all the polymorphisms associated with sporadic breast cancer are not associated with familial breast cancer.
Literature
1.
go back to reference Ali-Osman F, Akande O, Antoun G et al (1997) Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins. J Biol Chem 272:10004–10012PubMedCrossRef Ali-Osman F, Akande O, Antoun G et al (1997) Molecular cloning, characterization, and expression in Escherichia coli of full-length cDNAs of three human glutathione S-transferase Pi gene variants. Evidence for differential catalytic activity of the encoded proteins. J Biol Chem 272:10004–10012PubMedCrossRef
2.
go back to reference Ambrosone CB, Freudenheim JL, Graham S et al (1995) Cytochrome P4501A1 and glutathione S-transferase (M1) genetic polymorphisms and postmenopausal breast cancer risk. Cancer Res 55:3483–3485PubMed Ambrosone CB, Freudenheim JL, Graham S et al (1995) Cytochrome P4501A1 and glutathione S-transferase (M1) genetic polymorphisms and postmenopausal breast cancer risk. Cancer Res 55:3483–3485PubMed
3.
go back to reference Ambrosone CB, Sweeney C, Coles BF et al (2001) Polymorphisms in glutathione S-transferases (GSTM1 and GSTT1) and survival after treatment for breast cancer. Cancer Res 19:7130–7135 Ambrosone CB, Sweeney C, Coles BF et al (2001) Polymorphisms in glutathione S-transferases (GSTM1 and GSTT1) and survival after treatment for breast cancer. Cancer Res 19:7130–7135
4.
go back to reference Bell DA, Taylor JA, Paulson DF et al (1993) Genetic risk and carcinogen exposure: a common inherited defect of the carcinogen-metabolism gene glutathione S-transferase M1 (GSTM1) that increases susceptibility to bladder cancer. J Natl Cancer Inst 85:1159–1164PubMedCrossRef Bell DA, Taylor JA, Paulson DF et al (1993) Genetic risk and carcinogen exposure: a common inherited defect of the carcinogen-metabolism gene glutathione S-transferase M1 (GSTM1) that increases susceptibility to bladder cancer. J Natl Cancer Inst 85:1159–1164PubMedCrossRef
5.
go back to reference Chacko P, Joseph T, Mathew BS et al (2005) Role of xenobiotic metabolizing gene polymorphisms in breast cancer susceptibility and treatment outcome. Mutat Res 581:153–163PubMed Chacko P, Joseph T, Mathew BS et al (2005) Role of xenobiotic metabolizing gene polymorphisms in breast cancer susceptibility and treatment outcome. Mutat Res 581:153–163PubMed
6.
go back to reference Chen CL, Liu Q, Relling MV (1996) Simultaneous characterization of glutathione S-transferase M1 and T1 polymorphisms by polymerase chain reaction in American whites and blacks. Pharmacogenetics 6:187–191PubMedCrossRef Chen CL, Liu Q, Relling MV (1996) Simultaneous characterization of glutathione S-transferase M1 and T1 polymorphisms by polymerase chain reaction in American whites and blacks. Pharmacogenetics 6:187–191PubMedCrossRef
7.
go back to reference Curran JE, Weinstein SR, Griffiths LR (2000) Polymorphisms of glutathione S-transferase genes (GSTM1, GSTP1 and GSTT1) and breast cancer susceptibility. Cancer Lett 153:113–120PubMedCrossRef Curran JE, Weinstein SR, Griffiths LR (2000) Polymorphisms of glutathione S-transferase genes (GSTM1, GSTP1 and GSTT1) and breast cancer susceptibility. Cancer Lett 153:113–120PubMedCrossRef
8.
go back to reference DeMichele A, Aplenc R, Botbyl J et al (2005) Drug-metabolizing enzyme polymorphisms predict clinical outcome in a node-positive breast cancer cohort. J Clin Oncol 23:5552–5559PubMedCrossRef DeMichele A, Aplenc R, Botbyl J et al (2005) Drug-metabolizing enzyme polymorphisms predict clinical outcome in a node-positive breast cancer cohort. J Clin Oncol 23:5552–5559PubMedCrossRef
9.
go back to reference Easton DF (1999) How many more breast cancer predisposition genes are there? Breast Cancer Res 1:14–17PubMedCrossRef Easton DF (1999) How many more breast cancer predisposition genes are there? Breast Cancer Res 1:14–17PubMedCrossRef
10.
go back to reference Egan KM, Cai Q, Shu XO et al (2004) Genetic polymorphisms in GSTM1, GSTP1, and GSTT1 and the risk for breast cancer: results from the Shanghai Breast Cancer Study and meta-analysis. Cancer Epidemiol Biomarkers Prev 13:197–204PubMedCrossRef Egan KM, Cai Q, Shu XO et al (2004) Genetic polymorphisms in GSTM1, GSTP1, and GSTT1 and the risk for breast cancer: results from the Shanghai Breast Cancer Study and meta-analysis. Cancer Epidemiol Biomarkers Prev 13:197–204PubMedCrossRef
11.
go back to reference Ford D, Easton DF, Stratton M et al (1998) Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast cancer linkage consortium. Am J Hum Genet 62:676–689PubMedCrossRef Ford D, Easton DF, Stratton M et al (1998) Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast cancer linkage consortium. Am J Hum Genet 62:676–689PubMedCrossRef
12.
go back to reference Garcia-Closas M, Kelsey KT, Hankinson SE et al (1999) Glutathione S-transferase mu and theta polymorphisms and breast cancer susceptibility. J Natl Cancer Inst 91:1960–1964PubMedCrossRef Garcia-Closas M, Kelsey KT, Hankinson SE et al (1999) Glutathione S-transferase mu and theta polymorphisms and breast cancer susceptibility. J Natl Cancer Inst 91:1960–1964PubMedCrossRef
13.
go back to reference Gudmundsdottir K, Tryggvadottir L, Eyfjord JE (2001) GSTM1, GSTT1, and GSTP1 genotypes in relation to breast cancer risk and frequency of mutations in the p53 gene. Cancer Epidemiol Biomarkers Prev 10:1169–1173PubMed Gudmundsdottir K, Tryggvadottir L, Eyfjord JE (2001) GSTM1, GSTT1, and GSTP1 genotypes in relation to breast cancer risk and frequency of mutations in the p53 gene. Cancer Epidemiol Biomarkers Prev 10:1169–1173PubMed
14.
go back to reference Hamajima N, Takezaki T, Tajima K (2002) Allele Frequencies of 25 Polymorphisms pertaining to cancer risk for Japanese, Koreans and Chinese. Asian Pac J Cancer Prev 3:197–206PubMed Hamajima N, Takezaki T, Tajima K (2002) Allele Frequencies of 25 Polymorphisms pertaining to cancer risk for Japanese, Koreans and Chinese. Asian Pac J Cancer Prev 3:197–206PubMed
15.
go back to reference Harries LW, Stubbins MJ, Forman D et al (1997) Identification of genetic polymorphisms at the glutathione S-transferase Pi locus and association with susceptibility to bladder, testicular and prostate cancer. Carcinogenesis 18:641–644PubMedCrossRef Harries LW, Stubbins MJ, Forman D et al (1997) Identification of genetic polymorphisms at the glutathione S-transferase Pi locus and association with susceptibility to bladder, testicular and prostate cancer. Carcinogenesis 18:641–644PubMedCrossRef
16.
go back to reference Hayes JD, Pulford DJ (1995) The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Crit Rev Biochem Mol Biol 30:445–600PubMedCrossRef Hayes JD, Pulford DJ (1995) The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Crit Rev Biochem Mol Biol 30:445–600PubMedCrossRef
17.
go back to reference Helzlsouer KJ, Selmin O, Huang HY et al (1998) Association between glutathione S-transferase M1, P1, and T1 genetic polymorphisms and development of breast cancer. J Natl Cancer Inst 90:512–518PubMedCrossRef Helzlsouer KJ, Selmin O, Huang HY et al (1998) Association between glutathione S-transferase M1, P1, and T1 genetic polymorphisms and development of breast cancer. J Natl Cancer Inst 90:512–518PubMedCrossRef
18.
go back to reference IARC (1989) IARC Monographs on the evaluation of carcinogenic risks to humans, Vol 43, diesel and gasoline engine exhausts and some nitroarenes. Lyon, 41–185 IARC (1989) IARC Monographs on the evaluation of carcinogenic risks to humans, Vol 43, diesel and gasoline engine exhausts and some nitroarenes. Lyon, 41–185
19.
go back to reference Joseph T, Kusumakumary P, Chacko P et al (2004) Genetic polymorphism of CYP1A1, CYP2D6, GSTM1 and GSTT1 and susceptibility to acute lymphoblastic leukaemia in Indian children. Pediatr Blood Cancer 43:560–567PubMedCrossRef Joseph T, Kusumakumary P, Chacko P et al (2004) Genetic polymorphism of CYP1A1, CYP2D6, GSTM1 and GSTT1 and susceptibility to acute lymphoblastic leukaemia in Indian children. Pediatr Blood Cancer 43:560–567PubMedCrossRef
20.
go back to reference Kelsey KT, Hankinson SE, Colditz GA et al (1997) Glutathione S-transferase class mu deletion polymorphism and breast cancer: results from prevalent versus incident cases. Cancer Epidemiol Biomarkers Prev 6:511–515PubMed Kelsey KT, Hankinson SE, Colditz GA et al (1997) Glutathione S-transferase class mu deletion polymorphism and breast cancer: results from prevalent versus incident cases. Cancer Epidemiol Biomarkers Prev 6:511–515PubMed
21.
go back to reference Ketterer B (1998) Glutathione S-transferases and prevention of cellular free radical damage. Free Radic Res 28:647–658PubMedCrossRef Ketterer B (1998) Glutathione S-transferases and prevention of cellular free radical damage. Free Radic Res 28:647–658PubMedCrossRef
22.
go back to reference Khedhaier A, Remadi S, Corbex M et al (2003) Glutathione S-transferases (GSTT1 and GSTM1) gene deletions in Tunisians: susceptibility and prognostic implications in breast carcinoma. Br J Cancer 89:1502–1507PubMedCrossRef Khedhaier A, Remadi S, Corbex M et al (2003) Glutathione S-transferases (GSTT1 and GSTM1) gene deletions in Tunisians: susceptibility and prognostic implications in breast carcinoma. Br J Cancer 89:1502–1507PubMedCrossRef
23.
go back to reference Kim SU, Lee KM, Park SK et al (2004) Genetic polymorphism of glutathione S-transferase P1 and breast cancer risk. J Biochem Mol Biol 37:582–585PubMed Kim SU, Lee KM, Park SK et al (2004) Genetic polymorphism of glutathione S-transferase P1 and breast cancer risk. J Biochem Mol Biol 37:582–585PubMed
24.
go back to reference Lizard-Nacol S, Coudert B, Colosetti P et al (1999) Glutathione S-transferase M1 null genotype: lack of association with tumour characteristics and survival in advanced breast cancer. Breast Cancer Res 1:81–87PubMedCrossRef Lizard-Nacol S, Coudert B, Colosetti P et al (1999) Glutathione S-transferase M1 null genotype: lack of association with tumour characteristics and survival in advanced breast cancer. Breast Cancer Res 1:81–87PubMedCrossRef
25.
go back to reference Millikan R, Pittman G, Tse CK, Savitz DA, Newman B, Bell D (2000) Glutathione S-transferases M1, T1, and P1 and breast cancer. Cancer Epidemiol Biomarkers Prev 9:567–573PubMed Millikan R, Pittman G, Tse CK, Savitz DA, Newman B, Bell D (2000) Glutathione S-transferases M1, T1, and P1 and breast cancer. Cancer Epidemiol Biomarkers Prev 9:567–573PubMed
26.
go back to reference Mitrunen K, Jourenkova N, Kataja V et al (2001) Glutathione S-transferase M1, M3, P1, and T1 genetic polymorphisms and susceptibility to breast cancer. Cancer Epidemiol Biomarkers Prev 10:229–236PubMed Mitrunen K, Jourenkova N, Kataja V et al (2001) Glutathione S-transferase M1, M3, P1, and T1 genetic polymorphisms and susceptibility to breast cancer. Cancer Epidemiol Biomarkers Prev 10:229–236PubMed
27.
go back to reference Nathanson KL, Weber BL (2001) Other breast cancer susceptibility genes: searching for more holy grail. Hum Mol Genet 10:715–720PubMedCrossRef Nathanson KL, Weber BL (2001) Other breast cancer susceptibility genes: searching for more holy grail. Hum Mol Genet 10:715–720PubMedCrossRef
28.
go back to reference Nedelcheva KV, Andersen TI, Erikstein B et al (1998) Single tube multiplex polymerase chain reaction genotype analysis of GSTM1, GSTT1 and GSTP1: relation of genotypes to TP53 tumor status and clinicopathological variables in breast cancer patients. Pharmacogenetics 8:441–447CrossRef Nedelcheva KV, Andersen TI, Erikstein B et al (1998) Single tube multiplex polymerase chain reaction genotype analysis of GSTM1, GSTT1 and GSTP1: relation of genotypes to TP53 tumor status and clinicopathological variables in breast cancer patients. Pharmacogenetics 8:441–447CrossRef
29.
go back to reference Park SK, Yoo KY, Lee SJ et al (2000) Alcohol consumption, glutathione S-transferase M1 and T1 genetic polymorphisms and breast cancer risk. Pharmacogenetics 10:301–309PubMedCrossRef Park SK, Yoo KY, Lee SJ et al (2000) Alcohol consumption, glutathione S-transferase M1 and T1 genetic polymorphisms and breast cancer risk. Pharmacogenetics 10:301–309PubMedCrossRef
30.
go back to reference Pemble S, Schroeder KR, Spencer SR et al (1994) Human glutathione S-transferase theta (GSTT1): cDNA cloning and the characterization of a genetic polymorphism. Biochem J 300(Pt 1):271–276PubMed Pemble S, Schroeder KR, Spencer SR et al (1994) Human glutathione S-transferase theta (GSTT1): cDNA cloning and the characterization of a genetic polymorphism. Biochem J 300(Pt 1):271–276PubMed
31.
go back to reference Ryberg D, Skaug V, Hewer A et al (1997) Genotypes of glutathione transferase M1 and P1 and their significance for lung DNA adduct levels and cancer risk. Carcinogenesis 18:1285–1289PubMedCrossRef Ryberg D, Skaug V, Hewer A et al (1997) Genotypes of glutathione transferase M1 and P1 and their significance for lung DNA adduct levels and cancer risk. Carcinogenesis 18:1285–1289PubMedCrossRef
32.
go back to reference Samson M, Swaminathan R, Rama R et al (2007) Role of GSTM1 (Null/Present), GSTP1 (Ile105Val) and P53 (Arg72Pro) genetic polymorphisms and the risk of breast cancer: a case control study from South India. Asian Pac J Cancer Prev 8(2):253–257PubMed Samson M, Swaminathan R, Rama R et al (2007) Role of GSTM1 (Null/Present), GSTP1 (Ile105Val) and P53 (Arg72Pro) genetic polymorphisms and the risk of breast cancer: a case control study from South India. Asian Pac J Cancer Prev 8(2):253–257PubMed
33.
go back to reference Seidegard J, Vorachek WR, Pero RW et al (1988) Hereditary differences in the expression of the human glutathione transferase active on trans-stilbene oxide are due to a gene deletion. Proc Natl Acad Sci USA 85:7293–7297PubMedCrossRef Seidegard J, Vorachek WR, Pero RW et al (1988) Hereditary differences in the expression of the human glutathione transferase active on trans-stilbene oxide are due to a gene deletion. Proc Natl Acad Sci USA 85:7293–7297PubMedCrossRef
34.
go back to reference Serova OM, Mazoyer S, Puget N et al (1997) Mutations in BRCA1 and BRCA2 in breast cancer families: are there more breast cancer-susceptibility genes? Am J Hum Genet 60:486–495PubMed Serova OM, Mazoyer S, Puget N et al (1997) Mutations in BRCA1 and BRCA2 in breast cancer families: are there more breast cancer-susceptibility genes? Am J Hum Genet 60:486–495PubMed
35.
go back to reference Sreeja L, Syamala V, Hariharan S et al (2005) Possible risk modification by CYP1A1, GSTM1 and GSTT1 gene polymorphisms in lung cancer susceptibility in a South Indian population. J Hum Genet 50:618–627PubMedCrossRef Sreeja L, Syamala V, Hariharan S et al (2005) Possible risk modification by CYP1A1, GSTM1 and GSTT1 gene polymorphisms in lung cancer susceptibility in a South Indian population. J Hum Genet 50:618–627PubMedCrossRef
36.
go back to reference Sreelekha TT, Ramadas K, Pandey M et al (2001) Genetic polymorphism of CYP1A1, GSTM1 and GSTT1 genes in Indian oral cancer. Oral Oncol 37:593–598PubMedCrossRef Sreelekha TT, Ramadas K, Pandey M et al (2001) Genetic polymorphism of CYP1A1, GSTM1 and GSTT1 genes in Indian oral cancer. Oral Oncol 37:593–598PubMedCrossRef
37.
go back to reference Strange RC, Spiteri MA, Ramachandran S et al (2001) Glutathione-S-transferase family of enzymes. Mutat Res 482:21–26PubMed Strange RC, Spiteri MA, Ramachandran S et al (2001) Glutathione-S-transferase family of enzymes. Mutat Res 482:21–26PubMed
38.
go back to reference Sundberg K, Johansson AS, Stenberg G et al (1998) Differences in the catalytic efficiencies of allelic variants of glutathione transferase P1–1 towards carcinogenic diol epoxides of polycyclic aromatic hydrocarbons. Carcinogenesis 19:433–436PubMedCrossRef Sundberg K, Johansson AS, Stenberg G et al (1998) Differences in the catalytic efficiencies of allelic variants of glutathione transferase P1–1 towards carcinogenic diol epoxides of polycyclic aromatic hydrocarbons. Carcinogenesis 19:433–436PubMedCrossRef
39.
go back to reference Sweeney C, Ambrosone CB, Joseph L et al (2003) Association between a glutathione S-transferase A1 promoter polymorphism and survival after breast cancer treatment. Int J Cancer 103:810–814PubMedCrossRef Sweeney C, Ambrosone CB, Joseph L et al (2003) Association between a glutathione S-transferase A1 promoter polymorphism and survival after breast cancer treatment. Int J Cancer 103:810–814PubMedCrossRef
40.
go back to reference Yang G, Shu XO, Ruan ZX et al (2005) Genetic polymorphisms in glutathione-S-transferase genes (GSTM1, GSTT1, GSTP1) and survival after chemotherapy for invasive breast carcinoma. Cancer 103:52–58PubMedCrossRef Yang G, Shu XO, Ruan ZX et al (2005) Genetic polymorphisms in glutathione-S-transferase genes (GSTM1, GSTT1, GSTP1) and survival after chemotherapy for invasive breast carcinoma. Cancer 103:52–58PubMedCrossRef
41.
go back to reference Zimniak P, Nanduri B, Pikula S et al (1994) Naturally occurring human glutathione S-transferase GSTP1–1 isoforms with isoleucine and valine in position 104 differ in enzymic properties. Eur J Biochem 224:893–899PubMedCrossRef Zimniak P, Nanduri B, Pikula S et al (1994) Naturally occurring human glutathione S-transferase GSTP1–1 isoforms with isoleucine and valine in position 104 differ in enzymic properties. Eur J Biochem 224:893–899PubMedCrossRef
Metadata
Title
Influence of germline polymorphisms of GSTT1, GSTM1, and GSTP1 in familial versus sporadic breast cancer susceptibility and survival
Authors
Volga S. Syamala
Leelakumari Sreeja
Vani Syamala
Praveenkumar B. Raveendran
Rajan Balakrishnan
Ratheesan Kuttan
Ravindran Ankathil
Publication date
01-09-2008
Publisher
Springer Netherlands
Published in
Familial Cancer / Issue 3/2008
Print ISSN: 1389-9600
Electronic ISSN: 1573-7292
DOI
https://doi.org/10.1007/s10689-007-9177-1

Other articles of this Issue 3/2008

Familial Cancer 3/2008 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine