Next Article in Journal
Isolation and Molecular Characterization of 1-Aminocyclopropane-1-carboxylic Acid Synthase Genes in Hevea brasiliensis
Next Article in Special Issue
Non-Coding RNAs in Saliva: Emerging Biomarkers for Molecular Diagnostics
Previous Article in Journal
The Actin Depolymerizing Factor (ADF)/Cofilin Signaling Pathway and DNA Damage Responses in Cancer
Previous Article in Special Issue
Detection of miRNA as Non-Invasive Biomarkers of Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genetic Polymorphisms in Estrogen-Related Genes and the Risk of Breast Cancer among Han Chinese Women

1
Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
2
School of Biotechnology, Southern Medical University, Guangzhou 510515, China
3
Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research, Barcelona 08908, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2015, 16(2), 4121-4135; https://doi.org/10.3390/ijms16024121
Submission received: 3 November 2014 / Revised: 21 January 2015 / Accepted: 4 February 2015 / Published: 13 February 2015
(This article belongs to the Special Issue Emerging Classes of Biomarkers for Molecular Diagnostics)

Abstract

:
Exposure to high levels of estrogen is considered an important risk factor for susceptibility to breast cancer. Common polymorphisms in genes that affect estrogen levels may be associated with breast cancer risk, but no comprehensive study has been performed among Han Chinese women. In the present study, 32 single-nucleotide polymorphisms (SNPs) in estrogen-related genes were genotyped using the MassARRAY IPLEX platform in 1076 Han Chinese women. Genotypic and allelic frequencies were compared between case and control groups. Unconditional logistic regression was used to assess the effects of SNPs on breast cancer risk. Associations were also evaluated for breast cancer subtypes stratified by estrogen receptor (ER) and progesterone receptor (PR) status. Case-control analysis showed a significant relation between heterozygous genotypes of rs700519 and rs2069522 and breast cancer risk (OR = 0.723, 95% CI = 0.541–0.965, p = 0.028 and OR = 1.500, 95% CI = 1.078–2.087, p = 0.016, respectively). Subgroup comparisons revealed that rs2446405 and rs17268974 were related to ER status, and rs130021 was associated with PR status. Our findings suggest that rs700519 and rs2069522 are associated with susceptibility to breast cancer among the Han Chinese population and have a cumulative effect with three other identified SNPs. Further genetic and functional studies are needed to identify additional SNPs, and to elucidate the underlying molecular mechanisms.

1. Introduction

Breast cancer is one of the most common malignancies in women, and it causes serious health issues in women worldwide [1]. Although the incidence rate of breast cancer is currently low in China compared to Western countries, it is increasing rapidly. If current trends continue, the incidence rate in China will rise to approximately 85 per 100,000 woman-years by 2021 [2]. It is widely recognized that breast cancer development is the consequence of complex interactions between the genome, lifestyle, and environment. Many factors including family history, high mammographic density, postmenopausal hormone use, early age of menarche, late first pregnancy, breastfeeding for short periods, lower parity, and a longer interval between births have been confirmed to be associated with breast cancer susceptibility [3]. In addition, inheritance and hormones have also been suggested to be important risk factors [4,5].
The normal growth and function of breast epithelial cells are under hormonal control. Estrogen acts as a key driver for breast cancer because it can increase the frequency of mitotic activity, thereby stimulating cell divisions and increasing the potential of cells to accumulate mutations [6,7]. It is well established that high levels of estrogen or prolonged exposure is associated with breast cancer susceptibility [8]. A reanalysis of nine prospective studies demonstrated that there was a statistically significant increase in breast cancer risk with increasing concentrations of endogenous sex hormones in postmenopausal women [9]. Thus, the proper level of endogenous estrogen is crucial for female health. The biosynthesis and metabolism of estrogens, the main source of endogenous estrogens, is a complex process. A series of enzymes, such as cytochrome P450, family 17 (CYP17), CYP11A, CYP19, hydroxysteroid (17-β) dehydrogenase 1 (HSD17B1), CYP1A1, CYP1A2, CYP1B1, catechol-O-methyltransferase (COMT), as well as many other estrogen-signaling regulatory enzymes, is involved in the catabolism of cholesterol into estrogens and their subsequent degradation [10]. To exert their physiological effects, estrogens must first bind to estrogen receptors (ERs) and form complexes that subsequently bind to specific sequences in the promoters of estrogen-responsive genes [11]. Thus, all of these estrogen-related genes have been speculated to influence the synthesis or degradation of estrogens and are considered as candidate breast cancer susceptibility genes.
Single-nucleotide polymorphisms (SNPs) are the most frequent type of variation in the human genome, and are also valuable in tests of association with common diseases and pharmacogenetic traits [12,13]. Many studies have evaluated the association between SNPs in estrogen-related genes, such as CYP17, CYP19, CYP1A1, CYP1B1, COMT, GSTP1, and ERα/β, and the risk of breast cancer, especially in postmenopausal women [14,15,16,17,18,19,20,21]. Polymorphisms in these genes have been reported to affect the regulation, transcription, and activity of the enzymes, as well as subsequent exposure to endogenous estrogen and the development of breast cancer [14]. Although the effect of individual SNPs is usually small, multi-SNP association analysis can reveal cumulative risk effects and significantly relate to breast cancer [22]. Except for some reviews in which data from different reports and races were summarized and synthetically analyzed, most studies to date have focused on Western women [14,15]. Chinese populations have only been analyzed through studies with small sample sizes where few genes in the estrogen pathways were included. These conditions are too limited to explore and demonstrate special genes or SNPs of breast cancer genes in Han Chinese women comprehensively.
In a previous study, we genotyped six SNPs in the estrogen biosynthesis gene CYP11A1, and three variants (rs2279357, rs2959003, and rs2959008) were identified as associated with susceptibility to breast cancer [23]. In the present study, we explored the role of genetic polymorphisms in additional estrogen-related genes in breast cancer risk to systematically identify additional risk polymorphisms. We selected 32 SNPs, including 14 located in estrogen biosynthesis genes (CYP17A1, HSD17B1, CYP19A1, and STS), seven in metabolism genes (CYP1A1, CYP1A2, CYP1B1, COMT, and GSTP1), six in estrogen receptors (ERα and ERβ), and five in estrogen-signaling regulatory enzymes (CARM1, CREBBP, NQO1, SRD5A2, and SΜLT1E1). The study was performed on Guangdong Han Chinese women.

2. Results

2.1. Subject Characteristics and Quality Control of SNPs

The baseline characteristics of the 1076 subjects (546 controls and 530 cases) are shown in Table 1. The mean ages at the time of recruitment of the patients and controls significantly differed (48.48 ± 10.18 and 44.59 ± 11.26 years, respectively, p < 0.05); therefore, statistical analysis based on case-control comparisons was adjusted for age. The most common tumor histopathological type was invasive ductal carcinoma (IDC), accounting for 83.96% (445) compared to 4.91% (26) of other tumor types including invasive lobular carcinoma (ILC), mucin-producing carcinomas (Muc), and medullary carcinoma (Medul) and 11.13% (59) of unknown types. Only five patients (0.94%) presented with stage 0 (carcinoma in situ), and stages 1–4 accounted for 23.77%, 24.34%, 30.94%, and 8.87% of patients, respectively. The cases consisted of 270 (50.94%) ER-positive tumors, 190 (35.85%) ER-negative tumors, 237 (44.72%) PR-positive tumors, and 223 (42.08%) PR-negative tumors, whereas the remaining 70 tumors (13.21%) were untested.
Quality control of all of the SNPs, consisting of a Hardy-Weinberg equilibrium (HWE) p > 0.05 in healthy controls, a genotyping rate ≥ 85%, and a minor allele frequency (MAF) > 0.01 was conducted. Five SNPs (rs2830, rs4680, rs707762, rs17268974, and rs1695) that deviated from HWE were omitted from further case-control analysis.

2.2. Polymorphisms in Estrogen Biosynthesis Genes and Breast Cancer

We compared the genotypic and allelic frequencies of 11 SNPs in estrogen biosynthesis genes based on the case-control study and observed that rs700519 in CYP19A1 was significantly associated with breast cancer risk. The frequency of the heterozygous genotype C/T was lower in the breast cancer group than in the control group (20.9% vs. 26.2%). Women who carried the C/T genotype and T allele could be protected against breast cancer (OR = 0.723, 95% CI = 0.541–0.965, p = 0.028 and OR = 0.772, 95% CI = 0.600–0.993, p = 0.044, respectively). The results are shown in Table 2. No significant association was identified for the other SNPs.
Table 1. Baseline characteristics of the study subjects.
Table 1. Baseline characteristics of the study subjects.
VariableControl (n = 546)Case (n = 530)
EthnicityHan Chinese womenHan Chinese women
ResidenceGuangdong, ChinaGuangdong, China
Mean age ± SD (years) *44.59 ± 11.2648.48 ± 10.18
Mean age of menarche ± SD (years)14.50 ± 1.5414.33 ± 1.62
Mean BMI ± SD (kg/m2)23.33 ± 3.5823.57 ± 3.34
Exogenous hormone use a No411 (75.27%)373 (70.38%)
Yes107 (19.60%)114 (21.51%)
Unknown28 (5.13%)43 (8.11%)
No. full-term pregnancies Nulliparous25 (4.58%)37 (6.98%)
1211 (38.64%)214 (40.38%)
2184 (33.70%)172 (32.45%)
383 (15.20%)66 (12.45%)
≥443 (7.88%)41 (7.74%)
Variable in breast cancer cases
Family history Yes7 (1.32%)
No523 (98.68%)
Tumor histopathology IDC445 (83.96%)
Others b26 (4.91%)
Unknown59 (11.13%)
Clinical stage (UICC) c Stage 0 (in situ)5 (0.94%)
Stage 1126 (23.77%)
Stage 2129 (24.34%)
Stage 3164 (30.94%)
Stage 447 (8.87%)
Unknown59 (11.13%)
ER status Positive270 (50.94%)
Negative190 (35.85%)
Unknown70 (13.21%)
PR status Positive237 (44.72%)
Negative223 (42.08%)
Unknown70 (13.21%)
Abbreviation: BMI, body mass index; * p <0.05; a Include either oral contraceptives (OC) or hormone replacement therapy (HRT); b Mean ILC, Muc and Medul; c mean International Union Against Cancer (UICC) stages; invasive ductal carcinoma (IDC); invasive lobular carcinoma (ILC); mucin-producing carcinomas (Muc); and medullary carcinoma (Medul); ER, estrogen receptor; PR, progesterone receptor.
Table 2. Single single-nucleotide polymorphisms (SNPs) in estrogen biosynthesis genes and association with breast cancer.
Table 2. Single single-nucleotide polymorphisms (SNPs) in estrogen biosynthesis genes and association with breast cancer.
GeneSNPsAllelesCodominant ModelptrendAdditive Model
Common/RareHeterozygote OR (95% CI) ap aHomozygote OR (95% CI) ap aRare Allele OR (95% CI) ap a
CYP17A1rs743572G/A1.287 (0.978–1.694)0.0721.081 (0.756–1.547)0.6690.5311.074 (0.902–1.278)0.424
HSD17B1rs615942G/T0.927 (0.694–1.237)0.6051.077 (0.760–1.526)0.6760.9051.031 (0.866–1.227)0.735
CYP19A1rs2414096G/A1.099 (0.833–1.451)0.5041.133 (0.787–1.630)0.5030.4411.065 (0.896–1.267)0.457
CYP19A1rs2445759G/T0.888 (0.359–2.193)0.7961.815 (0.153–21.572)0.6370.9451.038 (0.471–2.288)0.926
CYP19A1rs2446405T/A0.957 (0.712–1.286)0.7690.830 (0.579–1.188)0.3070.4580.919 (0.774–1.091)0.336
CYP19A1rs700519C/T0.723 (0.541–0.965)0.0280.845 (0.342–2.085)0.7150.0470.772 (0.600–0.993)0.044
CYP19A1rs10046T/C0.785 (0.587–1.051)0.1030.748 (0.527–1.064)0.1060.0740.866 (0.729–1.028)0.100
CYP19A1rs749292G/A0.994 (0.746–1.326)0.9701.100 (0.777–1.557)0.5930.6271.045 (0.880–1.241)0.618
CYP19A1rs936306G/A0.869 (0.672–1.125)0.2870.869 (0.574–1.317)0.5080.3860.908 (0.755–1.092)0.305
CYP19A1rs1008805T/C0.990 (0.766–1.278)0.9361.138 (0.736–1.758)0.5610.7991.036 (0.860–1.248)0.712
CYP19A1rs11575899TCT/DEL1.029 (0.797–1.328)0.8260.954 (0.612–1.487)0.8370.8831.001 (0.830–1.207)0.992
Abbreviation: OR, Odds Ratio; 95% CI, 95% confidence interval; Significant values are marked in bold; a Adjusted by age.

2.3. Polymorphisms in Estrogen Metabolism Genes and Breast Cancer

Among the five SNPs in estrogen metabolism genes, the genotype distribution of CYP1A2 rs2069522 was significantly different between cases and controls. In contrast to rs700519, the heterozygote T/C of rs2069522 increased the risk of breast cancer (OR = 1.500, 95% CI = 1.078–2.087, p = 0.016, Table 3). The other four SNPs showed no evidence of an association with breast cancer.
Table 3. Single SNPs in estrogen metabolism genes and association with breast cancer.
Table 3. Single SNPs in estrogen metabolism genes and association with breast cancer.
GeneSNPsAllelesCodominant ModelptrendAdditive Model
Common/RareHeterozygote OR (95% CI) ap aHomozygote OR (95% CI) ap aRare Allele OR (95% CI) ap a
CYP1A1rs1531163A/G0.996 (0.762–1.303)0.9780.643 (0.283–1.459)0.2900.6180.938 (0.746–1.180)0.586
CYP1A1rs2606345C/A0.722 (0.479–1.088)0.1200.380 (0.034–4.241)0.4320.0620.709 (0.480–1.048)0.085
CYP1A2rs2069522T/C1.500 (1.078–2.087)0.0160.514 (0.127–2.081)0.3510.0911.305 (0.968–1.760)0.081
CYP1A2rs762551A/C0.876 (0.678–1.133)0.3130.873 (0.572–1.333)0.5290.3620.914 (0.761–1.099)0.339
CYP1B1rs1056836C/G1.073 (0.787–1.462)0.6570.715 (0.286–1.792)0.4750.9640.997 (0.761–1.306)0.985
Abbreviation: OR, Odds Ratio; 95% CI, 95% confidence interval. Significant values are marked in bold. a Adjusted by age.

2.4. Polymorphisms in Estrogen Receptors (ERs) and Regulatory Genes

No significant differences were found in genotypic or allelic distributions between the case and control groups, indicating that the 11 SNPs in the ERs and regulatory genes were not independently associated with breast cancer in our study population. Detailed information is shown in Table 4 and Table 5.
Table 4. Single SNPs in estrogen receptors and association with breast cancer.
Table 4. Single SNPs in estrogen receptors and association with breast cancer.
GeneSNPsAllelesCodominant ModelptrendAdditive Model
Common/RareHeterozygote OR (95% CI) ap aHomozygote OR (95% CI) ap aRare Allele OR (95% CI) ap a
ERαrs728524A/G1.137 (0.873–1.480)0.3400.628 (0.318–1.239)0.1800.9650.996 (0.801–1.238)0.972
ERαrs2234693T/C1.110 (0.839–1.469)0.4650.986 (0.663–1.465)0.9440.8921.022 (0.844–1.237)0.823
ERαrs9340799A/G0.945 (0.726–1.229)0.6711.222 (0.677–2.207)0.5050.9001.011 (0.818–1.250)0.918
ERαrs3798577T/C0.796 (0.605–1.048)0.1040.927 (0.654–1.314)0.6690.3780.936 (0.786–1.113)0.454
ERβrs1255998G/C0.929 (0.712–1.212)0.5850.979 (0.683–1.404)0.9080.7830.976 (0.818–1.164)0.783
ERβrs1256049G/A0.951 (0.725–1.247)0.7161.075 (0.753–1.533)0.6920.9251.022 (0.858–1.217)0.805
Abbreviation: OR, Odds Ratio; 95% CI, 95% confidence interval; a Adjusted by age.
Table 5. Single SNPs in estrogen-signaling regulatory enzymes and association with breast cancer.
Table 5. Single SNPs in estrogen-signaling regulatory enzymes and association with breast cancer.
GeneSNPsAllelesCodominant ModelptrendAdditive Model
Common/RareHeterozygote OR (95% CI) ap aHomozygote OR (95% CI) ap aRare Allele OR (95% CI) ap a
CARM1rs12460421G/A1.272 (0.947–1.708)0.1101.024 (0.720–1.455)0.8960.9801.018 (0.857–1.209)0.841
CREBBPrs130021A/G1.150 (0.877–1.507)0.3120.994 (0.691–1.431)0.9760.9301.024 (0.859–1.220)0.792
NQO1rs1469908G/A1.142 (0.876–1.490)0.3260.889 (0.435–1.819)0.7480.3801.069 (0.856–1.335)0.555
SRD5A2rs523349G/C1.068 (0.810–1.409)0.6391.024 (0.719–1.458)0.8970.7421.019 (0.857–1.212)0.833
SΜLT1E1rs3736599G/A0.983 (0.761–1.270)0.8961.105 (0.717–1.703)0.6520.6681.024 (0.851–1.233)0.801
Abbreviation: OR, Odds Ratio; 95% CI, 95% confidence interval; a Adjusted by age.

2.5. Cumulative Risk Analysis

CYP11A1 encodes the key enzyme that catalyzes the initial and rate-limiting steps of steroid hormone synthesis. In our previous study, three SNPs, rs2279357, rs2959003, and rs2959008, in CYP11A1 were validated to be associated with breast cancer risk (OR = 1.558, 95% CI = 1.092–2.222, p = 0.014; OR = 0.668, 95% CI = 0.460–0.971, p = 0.034 and OR = 0.669, 95% CI = 0.452–0.991, p = 0.045) [23]. To investigate whether these significant risk polymorphisms had a cumulative effect, an integrated analysis including these five SNPs was performed to evaluate the cumulative risk for breast cancer (Table 6). We defined the genotypes C/C of rs700519, T/C of rs2069522, and T/T of rs2279357, rs2959003, and rs2959008 as risk genotypes to conduct statistical analysis. Consistent with our hypothesis, women who carried more risk genotypes had a higher susceptibility for developing breast cancer (ptrend = 0.030). Women with five high-risk genotypes had a statistically significant 1.286 times higher risk of developing breast cancer than controls (OR = 2.286, 95% CI = 1.187–4.399, p = 0.013).
The admixture maximum likelihood (AML) method was employed to evaluate the cumulative effect from multiple variants. As expected, four SNPs (rs2279357, rs2959008, rs2959003, and rs2069522) were significant at the 5% level (p = 0.02, 0.02, 0.03, and 0.04) after adjusting for age based on the trend test for association. The other SNP (rs700519) failed to reach this threshold (p = 0.08).
Table 6. Cumulative risk of five susceptibility SNPs for breast cancer.
Table 6. Cumulative risk of five susceptibility SNPs for breast cancer.
The Number of Risk GenotypeControlCaseOR (95% CI) ap a
1/5220 (46.8%)201 (41.4%)1-
2/599 (21.1%)110 (22.6%)1.223 (0.873–1.714)0.242
3/581 (17.2%)79 (16.3%)1.118 (0.772–1.619)0.556
4/555 (11.7%)65 (13.4%)1.327 (0.877–2.007)0.181
5/515 (3.2%)31 (6.4%)2.286 (1.187–4.399)0.013
The five SNPs include rs700519, rs2069522, rs2279357, rs2959003, and rs2959008. Significant values are marked in bold; a Adjusted by age.
The global test yielded a marginally significant association for the whole estrogen-related pathway (pglobal = 0.047). Dividing into four functional sub-pathways for the global test revealed strong associations with the estrogen biosynthesis (pglobal = 0.026) and metabolism (pglobal = 0.014) sub-pathways. However, the other two sub-pathways showed no association with breast cancer.

2.6. Stratified Analysis Based on ER and Progesterone Receptor (PR) Status

Given the significance of hormone receptors in breast cancer progression and clinical diagnosis, stratified analysis, based on ER and PR status and genetic polymorphisms, was performed. Finally, three SNPs (rs2446405, rs17268974, and rs130021) were found to be distributed differently among breast cancer subgroups. Genotype A/A and allele A of rs2446405 showed a stronger association with ER(+) tumors than ER(−) (OR = 2.072, 95% CI = 1.147–3.742, p = 0.016 and OR = 1.341, 95% CI = 1.030–1.746, p = 0.030); however, for rs17268974, A/A was associated with ER(−) tumors (OR = 0.468, 95% CI = 0.243–0.900, p = 0.023). A/G carriers of rs130021 also showed a tendency toward PR(+) tumors (OR = 1.756, 95% CI = 1.164–2.648, p = 0.007). The complete results for the different inheritance models are shown in Table 7.
Table 7. Case-case comparisons for the associations of polymorphisms by subgroups of ER and PR status.
Table 7. Case-case comparisons for the associations of polymorphisms by subgroups of ER and PR status.
SubgroupsSNPsAllelesCodominant ModelptrendAdditive Model
Common/RareHeterozygote OR (95% CI)pHomozygote OR (95% CI)pRare Allele OR (95% CI)p
ER statusrs2446405T/A1.217 (0.779–1.900)0.3882.072 (1.147–3.742)0.0160.0191.341 (1.030–1.746)0.030
ER statusrs17268974T/A0.857 (0.564–1.303)0.4710.468 (0.243–0.900)0.0230.0430.782 (0.598–1.022)0.072
PR statusrs130021A/G1.756 (1.164–2.648)0.0071.188 (0.680–2.076)0.5450.2051.193 (0.915–1.556)0.192
Abbreviation: ER, estrogen receptor; PR, progesterone receptor; Significant values are marked in bold.

3. Discussion

In the present retrospective study, two SNPs, rs700519 and rs2069522, were identified as being associated with breast cancer risk. The non-synonymous coding SNP rs700519 (Arg264Cys) is located in exon 7 of CYP19A1, which encodes aromatase, the key enzyme in estrogen biosynthesis. Aromatase catalyzes the rate-limiting step in the conversion of testosterone and androstenedione to estradiol and estrone in a wide variety of tissues, including the ovary, placenta, brain, and adipose tissue [24]. The up-regulation of CYP19A1 expression has been observed to contribute to breast cancer [25]. Individual genetic polymorphisms in this gene have also previously been reported to be associated with enzymatic activity (C1123T and rs28757184), with certain pathologic phenotypes including breast cancer (Trp39Arg), and with poor efficacy of cancer treatment (rs4646) [26,27,28,29,30]. Studies in Chinese women have identified that the common missense polymorphism rs700519 alone or combined with other polymorphisms could significantly modulate the risk of endometriosis [31], polycystic ovary syndrome [32], and survival of breast cancer [33]. In the present population, this SNP was significantly associated with susceptibility to breast cancer, consistent with a result based on North Indian women [18]. However, some studies showed no significant association [34,35,36]. Another key enzyme of the pathway is CYP1A2, which also plays a crucial role in estrogen metabolism. Genetic variants of CYP1A2 have been shown to contribute to the risk of lung and breast cancer by interacting with environmental factors and drug metabolism through regulation of enzyme activity [37,38,39]. SNP rs2069522 is located at −2847 bp of CYP1A2, within a putative region that bidirectionally regulates the transcriptional activation of both CYP1A1 and CYP1A2 genes [40]. Despite the important function of rs2069522, its association with breast cancer risk has not been clearly demonstrated. Genetic studies have only shown that it had the potential to affect colorectal cancer risk and the treatment response to clozapine in schizophrenic patients among British Caucasians and Koreans, respectively [41,42]. However, those results were not significant after correction for multiple testing. Our study revealed for the first time that heterozygosity of rs2069522 significantly increased breast cancer risk. It is possible that this variant may influence the transcription and thus activity of CYP1A1 protein. It is particularly worth noting that the association of rs700519 and rs2069522 was only observed for heterozygotes, but not for homozygotes. One possibility is that the number of rare homozygotes for these two SNPs is too small to show significant evidence, which is consistent with the HapMap database and other reports [41,43]. Furthermore, the biological function of these two SNPs has not been clarified fully. Thus, further analyses based on a larger set of SNPs should be conducted to perform fine mapping of the locus, and to explore their biological functions and mechanisms in more detail.
Genetic polymorphisms in estrogen-related genes have been intensively investigated in many studies. However, these studies produced inconsistent results [15,16,17,18,21,31,44,45]. Several polymorphisms that were previously reported to contribute to breast cancer risk showed no significant association in our case-control analysis, including rs743572, rs1056836, rs1695, and rs2234693 [18,21,44,45]. The inconsistency might mainly be due to racial differences in susceptibility to breast cancer [46], because these studies were performed on various populations and subject numbers. For instance, variants in CYP1B1 (rs1056836), COMT (rs4680), and ERα (rs9340799) were significantly related to breast cancer susceptibility among Caucasian women [20,21], but we failed to find the same associations in our study population. Thus, it is necessary to replicate these risk loci to validate specific variants for Han Chinese women.
Polymorphisms in genes could affect the levels of transcription, translation, and activity of enzymes and result in wide inter-individual variability of responses to carcinogenic substances. Although the effect of a single polymorphism on tumorigenesis is weak, multiple loci could combine to generate a cumulative risk that cooperates with environmental exposure [47]. We have previously investigated the polymorphisms in CYP11A1 and found that rs2279357, rs2959003, and rs2959008 were related to breast cancer risk [23]. Combined with two associated variants in the present study, the cumulative analysis of these five verified variants revealed a much higher risk than a single SNP, a finding that agrees remarkably well with our expectation. The pathway-based AML global test also showed a significant association for the whole estrogen-related pathway that was much stronger for estrogen biosynthesis and metabolism sub-pathways. These findings support the multigenic nature of the etiology of breast cancer and need for gene–gene or locus–locus interactive studies on low-penetrance genes to comprehensively explore the underlying mechanisms of breast cancer.
It is necessary for all hormones to combine with their corresponding receptors to exert their biological functions. The expression of receptors, such as ER, PR, and human epidermal growth factor receptor-2 (HER2), is included in routine clinical classification and evaluation of breast cancer [48]. Their expression may also predict the response to targeted therapies such as tamoxifen and trastuzumab. Therefore, we also carried out stratified analyses based on ER and PR subtypes in the case group. Three SNPs (rs2446405, rs17268974, and rs130021) were confirmed to be associated with ER or PR status. Genetic changes in the receptors may influence their activity and therefore the risk and subtype of breast cancer.
Our study had some limitations. First, menopausal status can modify a woman’s risk of developing breast cancer, and the incidence rate differs between premenopausal and postmenopausal women. In the current study, we did not evaluate the associations for postmenopausal and premenopausal women separately because information on menopausal status was not obtained from most of the participants. Second, the statistical power for all of the 1076 subjects was larger than 83% to detect a log-additive genotype relative risk of 1.30 (Table S1). It was considered to be well powered to detect strong or moderate effect sizes of disease-predisposing variants, but smaller effects might have been missed. In fact, the exact powers of the two significant SNPs, rs700519 and rs2069522, were 68.12% and 83.69%, respectively. Furthermore, some variants in estrogen-related genes, such as CYP1A1 T3801C, CYP1A1 A2455G, and COMT Val158Met, were identified as susceptibility loci in breast cancer development according to recent meta-analysis reports [49,50,51]. However, these SNPs were not included in the present study because they failed in the assay design or deviated from HWE. It is worth noting that the significance of the results could be due to certain biases, which should be considered. Our study is still ongoing, and an additional large-scale assay with a more comprehensive design and more candidate SNPs may help to resolve these issues. It is, however, expected to improve the risk models for breast cancer among Han Chinese women.

4. Experimental Section

4.1. Ethics Statement

The present study was reviewed and approved by the regional ethical committee of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China (IRB No: 2009-SCBCGS-GZ-01) on 12 January 2009. Written informed consent was obtained from all of the participants in the trial.

4.2. Study Subjects

A total of 1076 Han Chinese women were included in the current study; 530 were cases with breast cancer and 546 were controls. All of the patients were recruited from Nanfang Hospital after a pathological diagnosis was ascertained. Clinical information was obtained from the patients’ medical records, including demographic characteristics, age at diagnosis, menopausal status, ER status, PR status, HER2 status, clinical stage, degree of differentiation, size of tumor, and lymph node involvement. The healthy controls were derived randomly and simultaneously from the same hospital, and were matched by ethnicity and geographic location. All of the controls were checked by a physician for the absence of cancer, and family history of their first-degree relatives was self-ascertained.

4.3. DNA Extraction

Peripheral blood samples were collected from the participants and stored at −70 °C until DNA extraction. Genomic DNA was extracted from EDTA-containing blood using the EZNA blood DNA kit (Omega Biotek, Norcross, GA, USA) according to the manufacturer’s instructions. The DNA was stored at −70 °C.

4.4. SNP Selection and Genotyping

The rationale for SNP selection has been described in detail elsewhere [23,52]. Briefly, we selected candidate SNPs in estrogen-related genes from Genome-Wide Association Studies GWAS, meta-analysis in multiple populations, or other large-sample association studies on breast cancer [41,44,53]. Furthermore, SNPs that were significantly associated with other hormone-induced cancers such as ovarian cancer and endometrial cancer were also selected. To achieve a power of at least 50%, with an odds ratio of 1.3, only SNPs with MAF ≥ 10% in the Chinese population according to the HapMap database were included. Monomorphic SNPs among the Chinese population were discarded. Finally, the genotyping assay was designed using Assay Design 4.0 software (Sequenom, San Diego, CA, USA), and after comprehensive evaluation, a total of 32 SNPs in 16 genes were included in this study. Details on the locations and characteristics of these SNPs are listed in Table S2.
For each SNP, a pair of amplification primers and an extension primer were designed using Assay Design 4.0 software. Genotyping was performed using the high-throughput MassARRAY iPLEX platform (Sequenom) through multiplex reactions. The genotyping rate of DNA samples was set to ≥85%.

4.5. Statistical Analysis

The statistical power of the case-control study was calculated using QUANTO software version 1.2.4 (University of Southern California, Los Angeles, CA, USA; http://hydra.usc.edu/gxe). We conducted a case-control study for all of the subjects, and then the patients were stratified by ER and PR status. Association analysis based on unconditional logistic regression was carried out by estimating OR and 95% CI with multiple inheritance models (codominant, dominant, recessive, overdominant, and additive) for each SNP. HWE was assessed using Fisher’s exact test and the χ2 test. The statistical tests were implemented using SPSS 13.0 software combined with the Web-based tool SNPstats (http://bioinfo.iconcologia.net/SNPstats). We also performed the AML-based global test of association for the whole estrogen-related pathway as well as for four sub-pathways after adjusting for age using software provided by the authors of Tyrer et al. [54]. All of the statistical analyses were two-tailed, and the significance level was set at a p value of 0.05.

5. Conclusions

In conclusion, we have presented an integrated analysis of the relationship between common variants in estrogen-related genes and breast cancer risk. Our study provides molecular genetic evidence suggesting that the independent and combined effects of these polymorphisms potentially reflect exposure to estrogen and as a consequence, an increase in breast cancer susceptibility. Our data suggest that this association could be specific to ER status. These findings may contribute to obtaining a more conclusive understanding about the underlying etiology of breast cancer and provide new strategies for breast cancer prevention, early detection, and specific treatment.

Supplementary Materials

Supplementary materials can be found at https://www.mdpi.com/1422-0067/16/02/4121/s1.

Acknowledgments

We thank all of the patients and healthy control subjects who participated in our study. This work was financially supported by the National High Technology Research and Development Program of China (Grant No. 2012AA020205), the National Natural Science Foundation of China (Grant No. 81302327 and Grant No.81401755), and China study abroad Scholarship (Grant No. 201308440152).

Author Contributions

Min-Ying Sun, Ming Li and Xue-Xi Yang conceived and designed the experiments; Min-Ying Sun and An-Na Zhu performed the experiments; Hong-Yan Du and Hui-Ying Liang analyzed the data; Fen-Xia Li and Ming Li contributed reagents/materials/analysis tools; Min-Ying Sun and Xue-Xi Yang wrote the paper; Gorka Ruiz de Garibay edited the paper for English language and style.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [PubMed]
  2. Ziegler, R.G.; Anderson, W.F.; Gail, M.H. Increasing breast cancer incidence in China: The numbers add up. J. Natl. Cancer Inst. 2008, 100, 1339–1341. [Google Scholar] [CrossRef] [PubMed]
  3. Colditz, G.A.; Rosner, B. Cumulative risk of breast cancer to age 70 years according to risk factor status: Data from the Nurses’ Health Study. Am. J. Epidemiol. 2000, 152, 950–964. [Google Scholar] [CrossRef] [PubMed]
  4. Hankinson, S.E.; Colditz, G.A.; Willett, W.C. Towards an integrated model for breast cancer etiology: The lifelong interplay of genes, lifestyle, and hormones. Breast Cancer Res. 2004, 6, 213–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Figueroa, J.D.; Brinton, L.A. Unraveling genes, hormones, and breast cancer. J. Natl. Cancer Inst. 2012, 104, 641–642. [Google Scholar] [CrossRef] [PubMed]
  6. Yager, J.D.; Davidson, N.E. Estrogen carcinogenesis in breast cancer. N. Engl. J. Med. 2006, 354, 270–282. [Google Scholar] [CrossRef] [PubMed]
  7. Yager, J.D. Endogenous estrogens as carcinogens through metabolic activation. J. Natl. Cancer Inst. Monogr. 2000, 27, 67–73. [Google Scholar] [CrossRef] [PubMed]
  8. Clemons, M.; Goss, P. Estrogen and the risk of breast cancer. N. Engl. J. Med. 2001, 344, 276–285. [Google Scholar] [CrossRef] [PubMed]
  9. Key, T.; Appleby, P.; Barnes, I.; Reeves, G. Endogenous sex hormones and breast cancer in postmenopausal women: Reanalysis of nine prospective studies. J. Natl. Cancer. Inst. 2002, 94, 606–616. [Google Scholar] [CrossRef] [PubMed]
  10. Kristensen, V.N.; Borresen-Dale, A.L. Molecular epidemiology of breast cancer: Genetic variation in steroid hormone metabolism. Mutat. Res. 2000, 462, 323–333. [Google Scholar] [CrossRef] [PubMed]
  11. Kumar, V.; Green, S.; Stack, G.; Berry, M.; Jin, J.R.; Chambon, P. Functional domains of the human estrogen receptor. Cell 1987, 51, 941–951. [Google Scholar] [CrossRef] [PubMed]
  12. Sachidanandam, R.; Weissman, D.; Schmidt, S.C.; Kakol, J.M.; Stein, L.D.; Marth, G.; Sherry, S.; Mullikin, J.C.; Mortimore, B.J.; Willey, D.L.; et al. A map of human genome sequence variation containing 1.42 million single nucleotide polymorphisms. Nature 2001, 409, 928–933. [Google Scholar] [CrossRef]
  13. Cooper, G.M.; Johnson, J.A.; Langaee, T.Y.; Feng, H.; Stanaway, I.B.; Schwarz, U.I.; Ritchie, M.D.; Stein, C.M.; Roden, D.M.; Smith, J.D.; et al. A genome-wide scan for common genetic variants with a large influence on warfarin maintenance dose. Blood 2008, 112, 1022–1027. [Google Scholar] [CrossRef]
  14. Mitrunen, K.; Hirvonen, A. Molecular epidemiology of sporadic breast cancer. The role of polymorphic genes involved in oestrogen biosynthesis and metabolism. Mutat. Res. 2003, 544, 9–41. [Google Scholar] [CrossRef] [PubMed]
  15. Miyoshi, Y.; Noguchi, S. Polymorphisms of estrogen synthesizing and metabolizing genes and breast cancer risk in Japanese women. Biomed. Pharmacother. 2003, 57, 471–481. [Google Scholar] [CrossRef] [PubMed]
  16. Huang, C.S.; Chern, H.D.; Chang, K.J.; Cheng, C.W.; Hsu, S.M.; Shen, C.Y. Breast cancer risk associated with genotype polymorphism of the estrogen-metabolizing genes CYP17, CYP1A1, and COMT: A multigenic study on cancer susceptibility. Cancer Res. 1999, 59, 4870–4875. [Google Scholar] [PubMed]
  17. Clendenen, T.; Zeleniuch-Jacquotte, A.; Wirgin, I.; Koenig, K.L.; Afanasyeva, Y.; Lundin, E.; Arslan, A.A.; Axelsson, T.; Försti, A.; Hallmans, G.; et al. Genetic variants in hormone-related genes and risk of breast cancer. PLoS One 2013, 8, e69367. [Google Scholar] [CrossRef]
  18. Chattopadhyay, S.; Siddiqui, S.; Akhtar, M.S.; Najm, M.Z.; Deo, S.V.; Shukla, N.K.; Husain, S.A. Genetic polymorphisms of ESR1, ESR2, CYP17A1, and CYP19A1 and the risk of breast cancer: A case control study from North India. Tumour Biol. 2014, 35, 4517–4527. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, L.; Gu, L.; Qian, B.; Hao, X.; Zhang, W.; Wei, Q.; Chen, K. Association of genetic polymorphisms of ER-alpha and the estradiol-synthesizing enzyme genes CYP17 and CYP19 with breast cancer risk in Chinese women. Breast Cancer Res. Treat. 2009, 114, 327–338. [Google Scholar] [CrossRef] [PubMed]
  20. Cerne, J.Z.; Pohar-Perme, M.; Novakovic, S.; Frkovic-Grazio, S.; Stegel, V.; Gersak, K. Combined effect of CYP1B1, COMT, GSTP1, and MnSOD genotypes and risk of postmenopausal breast cancer. J. Gynecol. Oncol. 2011, 22, 110–119. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, J.; Higuchi, R.; Modugno, F.; Li, J.; Umblas, N.; Lee, J.; Lui, L.Y.; Ziv, E.; Tice, J.A.; Cummings, S.R.; et al. Estrogen receptor α haplotypes and breast cancer risk in older Caucasian women. Breast Cancer Res. Treat. 2007, 106, 273–280. [Google Scholar] [CrossRef]
  22. Low, Y.L.; Li, Y.; Humphreys, K.; Thalamuthu, A.; Li, Y.; Darabi, H.; Wedrén, S.; Bonnard, C.; Czene, K.; Iles, M.M.; et al. Multi-variant pathway association analysis reveals the importance of genetic determinants of estrogenmetabolism in breast and endometrial cancer susceptibility. PLoS Genet. 2010, 6, e1001012. [Google Scholar] [CrossRef]
  23. Sun, M.; Yang, X.; Ye, C.; Xu, W.; Yao, G.; Chen, J.; Li, M. Risk-association of CYP11A1 polymorphisms and breast cancer among Han Chinese women in southern china. Int. J. Mol. Sci. 2012, 13, 4896–4905. [Google Scholar] [CrossRef] [PubMed]
  24. Simpson, E.R.; Mahendroo, M.S.; Means, G.D.; Kilgore, M.W.; Hinshelwood, M.M.; Graham-Lorence, S.; Amarneh, B.; Ito, Y.; Fisher, C.R.; Michael, M.D.; et al. Aromatase cytochrome P450, the enzyme responsible for estrogen biosynthesis. Endocr. Rev. 1994, 15, 342–355. [Google Scholar]
  25. Irahara, N.; Miyoshi, Y.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Quantitative analysis of aromatase mRNA expression derived from various promoters (I.4, I.3, PII and I.7) and its association with expression of TNF-α, IL-6 and COX-2 mRNAs in human breast cancer. Int. J. Cancer 2006, 118, 1915–1921. [Google Scholar] [CrossRef] [PubMed]
  26. Morishima, A.; Grumbach, M.M.; Simpson, E.R.; Fisher, C.; Qin, K. Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J. Clin. Endocrinol. Metab. 1995, 80, 3689–3698. [Google Scholar] [PubMed]
  27. Payne, E.J.; Ingley, E.; Dick, I.M.; Wilson, S.G.; Bond, C.S.; Prince, R.L. In vitro kinetic properties of the Thr201Met variant of human aromatase gene CYP19A1: Functional responses to substrate and product inhibition and enzyme inhibitors. J. Clin. Endocrinol. Metab. 2009, 94, 2998–3002. [Google Scholar] [CrossRef] [PubMed]
  28. Hirose, K.; Matsuo, K.; Toyama, T.; Iwata, H.; Hamajima, N.; Tajima, K. The CYP19 gene codon 39 Trp/Arg polymorphism increases breast cancer risk in subsets of premenopausal Japanese. Cancer Epidemiol. Biomark. Prev. 2004, 13, 1407–1411. [Google Scholar]
  29. Liu, L.; Bai, Y.X.; Zhou, J.H.; Sun, X.W.; Sui, H.; Zhang, W.J.; Yuan, H.H.; Xie, R.; Wei, X.L.; Zhang, T.T.; et al. A polymorphism at the 3'-UTR region of the aromatase gene is associated with the efficacy of the aromatase inhibitor, anastrozole, in metastatic breast carcinoma. Int. J. Mol. Sci. 2013, 14, 18973–18988. [Google Scholar] [CrossRef]
  30. Garcia-Casado, Z.; Guerrero-Zotano, A.; Llombart-Cussac, A.; Calatrava, A.; Fernandez-Serra, A.; Ruiz-Simon, A.; Gavila, J.; Climent, M.A.; Almenar, S.; Cervera-Deval, J.; et al. A polymorphism at the 3'-UTR region of the aromatase gene defines a subgroup of postmenopausal breast cancer patients with poor response to neoadjuvant letrozole. BMC Cancer 2010, 10, 36. [Google Scholar] [CrossRef]
  31. Wang, H.S.; Wu, H.M.; Cheng, B.H.; Yen, C.F.; Chang, P.Y.; Chao, A.; Lee, Y.S.; Huang, H.D.; Wang, T.H. Functional analyses of endometriosis-related polymorphisms in the estrogen synthesis and metabolism-related genes. PLoS One 2012, 7, e47374. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, H.; Li, Q.; Wang, T.; Yang, G.; Wang, Y.; Zhang, X.; Sang, Q.; Wang, H.; Zhao, X.; Xing, Q.; et al. A common polymorphism in the human aromatase gene alters the risk for polycystic ovary syndrome and modifies aromatase activity in vitro. Mol. Hum. Reprod. 2011, 17, 386–391. [Google Scholar] [CrossRef]
  33. Long, J.R.; Kataoka, N.; Shu, X.O.; Wen, W.; Gao, Y.T.; Cai, Q.; Zheng, W. Genetic polymorphisms of the CYP19A1 gene and breast cancer survival. Cancer Epidemiol. Biomark. Prev. 2006, 15, 2115–2122. [Google Scholar] [CrossRef]
  34. Ma, X.; Qi, X.; Chen, C.; Lin, H.; Xiong, H.; Li, Y.; Jiang, J. Association between CYP19 polymorphisms and breast cancer risk: Results from 10,592 cases and 11,720 controls. Breast Cancer Res. Treat. 2010, 122, 495–501. [Google Scholar] [CrossRef] [PubMed]
  35. Haiman, C.A.; Dossus, L.; Setiawan, V.W.; Stram, D.O.; Dunning, A.M.; Thomas, G.; Thun, M.J.; Albanes, D.; Altshuler, D.; Ardanaz, E.; et al. Genetic variation at the CYP19A1 locus predicts circulating estrogen levels but not breast cancer risk in postmenopausal women. Cancer Res. 2007, 67, 1893–1897. [Google Scholar] [CrossRef]
  36. Diergaarde, B.; Potter, J.D.; Jupe, E.R.; Manjeshwar, S.; Shimasaki, C.D.; Pugh, T.W.; Defreese, D.C.; Gramling, B.A.; Evans, I.; White, E. Polymorphisms in genes involved in sex hormone metabolism, estrogen plus progestin hormone therapy use, and risk of postmenopausal breast cancer. Cancer Epidemiol. Biomark. Prev. 2008, 17, 1751–1759. [Google Scholar] [CrossRef]
  37. Ma, Z.; Guo, W.; Gong, T.; Niu, H.J.; Wang, R.W.; Jiang, Y.G. CYP1A2 rs762551 polymorphism contributes to risk of lung cancer: A meta-analysis. Tumour Biol. 2014, 35, 2253–2257. [Google Scholar] [CrossRef] [PubMed]
  38. Ayari, I.; Fedeli, U.; Saguem, S.; Hidar, S.; Khlifi, S.; Pavanello, S. Role of CYP1A2 polymorphisms in breast cancer risk in women. Mol. Med. Rep. 2013, 7, 280–286. [Google Scholar] [PubMed]
  39. Altar, C.A.; Hornberger, J.; Shewade, A.; Cruz, V.; Garrison, J.; Mrazek, D. Clinical validity of cytochrome P450 metabolism and serotonin gene variants in psychiatric pharmacotherapy. Int. Rev. Psychiatry 2013, 25, 509–533. [Google Scholar] [CrossRef] [PubMed]
  40. Ueda, R.; Iketaki, H.; Nagata, K.; Kimura, S.; Gonzalez, F.J.; Kusano, K.; Yoshimura, T.; Yamazoe, Y. A common regulatory region functions bidirectionally in transcriptional activation of the human CYP1A1 and CYP1A2 genes. Mol. Pharmacol. 2006, 69, 1924–1930. [Google Scholar] [CrossRef] [PubMed]
  41. Bethke, L.; Webb, E.; Sellick, G.; Rudd, M.; Penegar, S.; Withey, L.; Qureshi, M.; Houlston, R. Polymorphisms in the cytochrome P450 genes CYP1A2, CYP1B1, CYP3A4, CYP3A5, CYP11A1, CYP17A1, CYP19A1 and colorectal cancer risk. BMC Cancer 2007, 7, 123. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, S.T.; Ryu, S.; Kim, S.R.; Kim, M.J.; Kim, S.; Kim, J.W.; Lee, S.Y.; Hong, K.S. Association study of 27 annotated genes for clozapine pharmacogenetics: Validation of preexisting studies and identification of a new candidate gene, ABCB1, for treatment response. J. Clin. Psychopharmacol. 2012, 32, 441–448. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, J.Z.; Deogan, M.S.; Lewis, J.R.; Chew, S.; Mullin, B.H.; McNab, T.J.; Wilson, S.G.; Ingley, E.; Prince, R.L. A non-synonymous coding change in the CYP19A1 gene Arg264Cys (rs700519) does not affect circulating estradiol, bone structure or fracture. BMC Med. Genet. 2011, 12, 165. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, B.; Beeghly-Fadiel, A.; Long, J.; Zheng, W. Genetic variants associated with breast-cancer risk: Comprehensive research synopsis, meta-analysis, and epidemiological evidence. Lancet Oncol. 2011, 12, 477–488. [Google Scholar] [CrossRef] [PubMed]
  45. Reding, K.W.; Weiss, N.S.; Chen, C.; Li, C.I.; Carlson, C.S.; Wilkerson, H.W.; Farin, F.M.; Thummel, K.E.; Daling, J.R.; Malone, K.E. Genetic polymorphisms in the catechol estrogen metabolism pathway and breast cancer risk. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1461–1467. [Google Scholar] [CrossRef]
  46. Reding, K.W.; Chen, C.; Lowe, K.; Doody, D.R.; Carlson, C.S.; Chen, C.T.; Houck, J.; Weiss, L.K.; Marchbanks, P.A.; Bernstein, L.; et al. Estrogen-related genes and their contribution to racial differences in breast cancer risk. Cancer Causes Control 2012, 23, 671–681. [Google Scholar] [CrossRef] [PubMed]
  47. Liyanarachchi, S.; Wojcicka, A.; Li, W.; Czetwertynska, M.; Stachlewska, E.; Nagy, R.; Hoag, K.; Wen, B.; Ploski, R.; Ringel, M.D.; et al. Cumulative risk impact of five genetic variants associated with papillary thyroid carcinoma. Thyroid 2013, 23, 1532–1540. [Google Scholar] [CrossRef] [PubMed]
  48. O’Brien, K.M.; Cole, S.R.; Engel, L.S.; Bensen, J.T.; Poole, C.; Herring, A.H.; Millikan, R.C. Breast cancer subtypes and previously established genetic risk factors: A bayesian approach. Cancer Epidemiol. Biomark. Prev. 2014, 23, 84–97. [Google Scholar] [CrossRef]
  49. He, X.F.; Wei, W.; Liu, Z.Z.; Shen, X.L.; Yang, X.B.; Wang, S.L.; Xie, D.L. Association between the CYP1A1 T3801C polymorphism and risk of cancer: Evidence from 268 case-control studies. Gene 2014, 534, 324–344. [Google Scholar] [CrossRef] [PubMed]
  50. Qin, J.; Zhang, J.X.; Li, X.P.; Wu, B.Q.; Chen, G.B.; He, X.F. Association between the CYP1A1 A2455G polymorphism and risk of cancer: Evidence from 272 case-control studies. Tumour Biol. 2014, 35, 3363–3376. [Google Scholar] [CrossRef] [PubMed]
  51. Li, K.; Li, W.; Zou, H. Catechol-O-methyltransferase Val158Met polymorphism and breast cancer risk in Asian population. Tumour Biol. 2014, 35, 2343–2350. [Google Scholar] [CrossRef] [PubMed]
  52. He, X.; Yao, G.; Li, F.; Li, M.; Yang, X. Risk-association of five SNPs in TOX3/LOC643714 with breast cancer in southern China. Int. J. Mol. Sci. 2014, 15, 2130–2141. [Google Scholar] [CrossRef] [PubMed]
  53. Zheng, W.; Zhang, B.; Cai, Q.; Sung, H.; Michailidou, K.; Shi, J.; Choi, J.Y.; Long, J.; Dennis, J.; Humphreys, M.K.; et al. Common genetic determinants of breast-cancer risk in East Asian women: A collaborative study of 23,637 breast cancer cases and 25,579 controls. Hum. Mol. Genet. 2013, 22, 2539–2550. [Google Scholar] [CrossRef] [PubMed]
  54. Tyrer, J.; Pharoah, P.D.; Easton, D.F. The admixture maximum likelihood test: A novel experiment-wise test of association between disease and multiple SNPs. Genet. Epidemiol. 2006, 30, 636–643. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Sun, M.-Y.; Du, H.-Y.; Zhu, A.-N.; Liang, H.-Y.; De Garibay, G.R.; Li, F.-X.; Li, M.; Yang, X.-X. Genetic Polymorphisms in Estrogen-Related Genes and the Risk of Breast Cancer among Han Chinese Women. Int. J. Mol. Sci. 2015, 16, 4121-4135. https://doi.org/10.3390/ijms16024121

AMA Style

Sun M-Y, Du H-Y, Zhu A-N, Liang H-Y, De Garibay GR, Li F-X, Li M, Yang X-X. Genetic Polymorphisms in Estrogen-Related Genes and the Risk of Breast Cancer among Han Chinese Women. International Journal of Molecular Sciences. 2015; 16(2):4121-4135. https://doi.org/10.3390/ijms16024121

Chicago/Turabian Style

Sun, Min-Ying, Hong-Yan Du, An-Na Zhu, Hui-Ying Liang, Gorka Ruiz De Garibay, Fen-Xia Li, Ming Li, and Xue-Xi Yang. 2015. "Genetic Polymorphisms in Estrogen-Related Genes and the Risk of Breast Cancer among Han Chinese Women" International Journal of Molecular Sciences 16, no. 2: 4121-4135. https://doi.org/10.3390/ijms16024121

Article Metrics

Back to TopTop