Back to Journals » OncoTargets and Therapy » Volume 10

Association of estrogen receptor α PvuII and XbaI polymorphisms with prostate cancer susceptibility and risk stratification: a meta-analysis from case-control studies

Authors Zhao YN, Zheng X, Zhang LJ, Hu Q, Guo YT, Jiang H, Shi SN, Zhang X 

Received 14 January 2017

Accepted for publication 29 March 2017

Published 29 June 2017 Volume 2017:10 Pages 3203—3210

DOI https://doi.org/10.2147/OTT.S132419

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Ingrid Espinoza



Yining Zhao,1,* Xi Zheng,2,* Lijie Zhang,3 Qiang Hu,3 Yitian Guo,3 Hua Jiang,3 Shennan Shi,4 Xiang Zhang1

1Department of Urology, Qilu Hospital of Shandong University, Jinan, 2Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 3Department of Urology, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, 4Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People’s Republic of China

*These authors contributed equally to this work

Background: Studies on the association between two single nucleotide polymorphisms (SNPs) in estrogen receptor α (ERα), PvuII (rs2234693 T>C) and XbaI (rs9340799 A>G), and the prostate cancer risk are inconsistent. Therefore, we performed a meta-analysis to derive a more accurate estimation of this relationship.
Methods: A literature search of PubMed, Embase, Web of Science databases until October 1, 2016, was conducted. Crude odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to assess the strength of this association.
Results: Eighteen case-control studies, with a total of 3,317 prostate cancer patients and 8,324 controls, were included. Results showed that both PvuII and XbaI polymorphisms were significantly associated with a higher prostate cancer risk in overall populations. To derive a more accurate estimation, subgroup analysis stratified by ethnicity revealed that this relationship existed only in Caucasians, but not in Asians. Furthermore, PvuII polymorphism was significantly associated with high Gleason grade (Gleason score ≥7) cancers.
Conclusion: The current meta-analysis demonstrates that ERα PvuII and XbaI polymorphisms are associated with a higher prostate cancer risk in Caucasians, but not in Asians, and PvuII polymorphism is significantly associated with high Gleason grade tumors, indicating the probability of inherited susceptibility to prostate cancer arising from different genomic ERα SNPs, which may help us understand the pathogenesis of prostate cancer in Caucasians.

Keywords: estrogen receptor α, PvuII, XbaI, prostate cancer, meta-analysis

Introduction

Prostate cancer is the most common malignancy in men and a major cause of cancer-related deaths.1 Since prostate-specific antigen (PSA)-based screening regime for prostate cancer remains controversial because of the high rate of overdiagnosis and overtreatment, a validated biomarker to complement PSA for screening and prognostic biomarkers with clinical utility remain an unmet need.2

In addition to androgens, estrogens also affect prostatic growth and carcinogenesis.3 The cellular effects of estrogens are mediated by two estrogen receptors (ERs), ERα and ERβ. The human ERα encoding gene locates on chromosome 6q25.1 and consists of eight exons and seven introns.4 Previous studies showed that the expression of ERα is gradually increased from prostate intraepithelial neoplasia, locally invasive cancers to metastatic lesions at both mRNA and protein levels.5 Furthermore, in vivo studies using ERα knockout mice revealed that ERα is an important determinant of prostatic carcinogenesis.3

Although several single nucleotide polymorphisms (SNPs) have been identified in the ERα gene, only a few have been extensively studied in prostate cancer. Furthermore, substantial controversial conclusions have been drawn by these studies. To address these issues, we chose two common polymorphisms in the ERα gene, PvuII (rs2234693 T>C) and XbaI (rs9340799 A>G), and conducted a meta-analysis on case-control studies between prostate cancer patients and prostate cancer-free controls. The aim of this study was to investigate the potential role of ERα PvuII and XbaI polymorphisms in the prostate cancer risk stratification.

Methods

The current meta-analysis was designed and reported according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement (Table S1).6

Literature search

Relevant papers published before October 1, 2016, were identified through a literature search in PubMed, Embase, and Cochrane databases using the following strategy: [(“gene mutation” OR “polymorphism”) OR (“SNP”) OR (“genetic variants”)] AND [(“prostate carcinoma”) OR (“prostate neoplasms”) OR (“prostate cancer”)] AND [(“estrogen receptor”) OR (“estradiol receptor”) OR (“ER”) OR (“ESR”)]. References from eligible articles were also manually searched to identify other potential publications.

Study eligibility

Studies were eligible for inclusion if, 1) studies focused on the association between ERα PvuII and XbaI polymorphisms and the prostate cancer risk; 2) they were case-control clinical studies on human subjects; 3) all diagnoses with prostate cancer were confirmed by pathological or histological examinations and controls were confirmed to be cancer free; 4) sufficient data were provided to estimate crude odds ratios (ORs) with 95% confidence intervals (CIs).

Two authors independently completed the screening process according to the Cochrane Collaboration guidelines.7 The methodological quality of each retrieved study was assessed using the Strengthening the Reporting of Genetic Association Studies (STREGA) quality score system.8 Forty-nine assessment items related to the quality appraisal were used in this score system with scores ranging from 0 to 49. Scores of 0–25, 26–37, and 38–49 were defined as low, moderate, and high quality, respectively (Table S2).

Data extraction

Detailed data from each publication were independently extracted by two authors using a standardized data extraction sheet and were checked by a third author (Table S3). The following information was extracted from each article: first author, year of publication, SNP ID and alternate name, ethnicity, country, language, study design, number of subjects, source of cases and controls, detecting sample, genotype method, allele and genotype frequencies, minor allele frequency (MAF), and evidence of Hardy–Weinberg equilibrium (HWE) in controls. Clinical parameters of prostate cancer patients were also collected, including tumor stage, serum PSA level at prostate cancer diagnosis, age at prostate cancer diagnosis, and smoking status. Risk of bias in each individual study was also evaluated.

Statistical analysis

The association strength between both polymorphisms and the prostate cancer risk was measured by ORs with 95% CIs under five genetic models: allele model, dominant model, recessive model, homozygous model, and heterozygous model. The statistical significance of the pooled OR was examined by the Z test. Heterogeneity between studies was estimated using the I2 test. I2<50% indicated that heterogeneity among studies was acceptable, and the fixed effects model (the Mantel–Haenszel method) was used. Otherwise, the random effects model (DerSimonian Laird method) was used. Subgroup analyses through ethnicity, country, source of cases and controls, genotype method, and whether HWE was in control or not were also conducted. We also stratified prostate cancer patients according to selected clinical parameters. HWE was evaluated by the χ2 test in controls. Sensitivity analysis was performed by omitting each study in order to assess the stability of pooled results. Begger’s funnel plots and Egger’s linear regression test were used to evaluate publication bias. If significant publication bias existed, the trim and fill method was used to adjust pooled estimates.9 All tests were two sided, and P<0.05 was considered statistically significant. Data analysis was performed using the STATA software (version 12.0; Stata Corp, College Station, TX, USA).

Results

Characteristics of included studies

According to the inclusion criteria, 18 case-control studies were included in the current meta-analysis.1027 The flow chart presenting the selection process is shown in Figure 1. A total of 3,317 prostate cancer patients and 8,324 controls were included in the synthesis. Years of publications ranged from 2001 to 2015. Genotype frequencies among controls were consistent with the HWE test, except for three studies.11,24,26 STREGA scores ranged from 29 to 36, which suggested that all of these studies were qualified to the quantitative synthesis. The characteristics and methodological quality of the included studies are summarized in Tables 1 and S4.

Figure 1 Flow of information through different phases of the present meta-analysis.
Abbreviation: SNP, single nucleotide polymorphisms.

Table 1 Characteristics and methodological quality of included studies
Notes: The study also used blood as testing sample. All cases came from hospital-based populations.
Abbreviations: SOC, source of controls; HWE, Hardy–Weinberg equilibrium (Y, HWE in controls; N, HWE out of controls); STREGA, Strengthening the Reporting of Genetic Association Studies; PB, population-based; PCR, polymerase chain reaction; NA, nonapplicable; HB, hospital-based.

ERα PvuII (rs2234693 T>C) polymorphism and the prostate cancer risk

Eighteen studies, comprising 3,317 cases and 8,324 controls, were involved in the synthesis. The results showed that PvuII polymorphism was related to an increased prostate cancer risk in overall populations under four genetic models (allele model OR: 1.16, 95% CI: 1.04−1.29, P<0.01; dominant model OR: 1.24, 95% CI: 1.06−1.47, P=0.01; recessive model OR: 1.16, 95% CI: 1.04−1.30, P<0.01; homozygous model OR: 1.37, 95% CI: 1.09−1.72, P<0.01; Tables 2 and S5). To derive a more accurate estimation, subgroup analyses stratified by ethnicity were conducted. The results indicated that PvuII polymorphism was significantly correlated with a higher prostate cancer risk in Caucasians (allele model OR: 1.12, 95% CI: 1.03−1.21, P<0.01; dominant model OR: 1.19, 95% CI: 1.05−1.35, P<0.01; recessive model OR: 1.13, 95% CI: 1.03−1.29, P=0.04; homozygous model OR: 1.25, 95% CI: 1.06−1.46, P=0.01; Tables 2 and S5), but not in Asians. Furthermore, when grouped according to Gleason grades, PvuII polymorphism was found to be significantly correlated with high Gleason grade cancers (Gleason score ≥7) under the allele model (OR: 1.90, 95% CI: 1.44−2.50, P<0.01, Table 3).

Table 2 Meta-analysis findings on the association between ERα PvuII (rs2234693 T>C) polymorphism and prostate cancer risk
Notes: If I2<50%, the fixed effects model was used. Otherwise, the random effects model was used.
Abbreviations: ERα, estrogen receptor α; OR, crude odds ratio; CI, confidence interval; HWE, Hardy–Weinberg equilibrium.

Table 3 Subgroup analysis by Gleason score of the association between ERα PvuII (rs2234693 T>C) polymorphism and prostate cancer risk under the allele model (T vs C)
Note: I2<50%, the fixed effects model was used.
Abbreviations: ERα, estrogen receptor α; OR, crude odds ratio; CI, confidence interval.

Sensitivity analysis suggested that individual studies did not affect pooled ORs (Figure S1). Begger’s funnel plot and Egger’s linear regression test did not show any statistical evidence of publication bias (Figure 2A). In addition, we excluded one study that deviated significantly from HWE, as the results showed that this non-HWE study had no effect on pooled ORs (Table 2).

Figure 2 Begg’s funnel plot for the publication bias test of the meta-analysis.
Notes: Each point represents a separate study for the indicated association. Horizontal line represents size of effect. (A) ERα PvuII polymorphism (t=1.07, P=0.30) and (B) ERα XbaI polymorphism (t=0.66, P=0.52).
Abbreviations: ERα, estrogen receptor α; OR, crude odds ratio; SE, standard error.

ERα XbaI (rs9340799 A>G) polymorphism and the prostate cancer risk

The correlation between XbaI polymorphism and the prostate cancer risk was investigated in 13 studies with 1,946 cases and 2,744 controls. The results showed that XbaI polymorphism had a positive association with the risk of prostate cancer in overall populations under four genetic models (allele model OR: 1.23, 95% CI: 1.06−1.43, P<0.01; dominant model OR: 1.38, 95% CI: 1.11−1.72, P<0.01; recessive model OR: 1.24, 95% CI: 1.03−1.49, P=0.03; homozygous model OR: 1.44, 95% CI: 1.17−1.76, P<0.01 Tables 4 and S6). Similarly, we performed a subgroup analyses based on ethnicity to derive a more accurate estimate, and the results showed that XbaI polymorphism was significantly correlated with an increased risk of prostate cancer in Caucasians (allele model OR: 1.31, 95% CI: 1.07−1.61, P<0.01; dominant model OR: 1.43, 95% CI: 1.07−1.92, P=0.01; recessive model OR: 1.36, 95% CI: 1.09−1.69, P<0.01; homozygous model OR: 1.48, 95% CI: 1.17−1.88, P<0.01; Tables 4 and S6), but not in Asians.

Table 4 Meta-analysis findings on the association between ERα XbaI (rs9340799 A>G) polymorphism and prostate cancer risk
Notes: If I2<50%, the fixed effects model was used. Otherwise, the random effects model was used.
Abbreviations: ERα, estrogen receptor α; OR, crude odds ratio; CI, confidence interval; HWE, Hardy–Weinberg equilibrium.

Sensitivity analysis suggested that individual studies did not affect pooled ORs (Figure S2). Begger’s funnel plot and Egger’s linear regression test did not show any statistical evidence of publication bias (Figure 2B). Moreover, we excluded two studies that deviated significantly from the HWE test, and the results showed that these two non-HWE studies had no effect on pooled ORs (Table 4).

Discussion

The role of PSA as a biomarker in prostate cancer remains unsatisfactory, leading to considerable overdiagnoses and overtreatment. This study on novel biomarkers in prostate cancer represents a long-standing hotspot in biomedical research.28 Since ERα is an important determinant of prostatic carcinogenesis,3 various studies have focused on SNPs in the ERα gene to determine their possible associations with the prostate cancer susceptibility. In the current study, we chose two common SNPs in the ERα gene, PvuII (rs2234693 T>C) and XbaI (rs9340799 A>G), and conducted a meta-analysis to evaluate their associations with prostate cancer susceptibility.

To our knowledge, the present work represents the largest meta-analysis performed to estimate the association of ERα SNPs with the risk of prostate cancer. The results showed that both PvuII and XbaI polymorphisms were correlated with an increased risk of prostate cancer in Caucasians, but not in Asians, and this disparity might be attributable to discrepancies in racial backgrounds and geography.14,29 Furthermore, there was a significant positive correlation of PvuII polymorphism and high Gleason grade cancers (Gleason score ≥7), which indicated its potential role in prostate cancer malignant transformation.

Both PvuII and XbaI polymorphisms lie in intron 1 of the ERα gene, which is part of the A/B domain, the transactivating factor 1. This domain is an important site for stimulating transcription from certain estrogen-responsive promoters.25 Among the possible explanations as to how these intronic polymorphisms affected prostate cancer risk are that, 1) both polymorphisms may be in linkage disequilibrium with other unknown variants in the gene, which may affect the gene expression or function;24,25,30 2) the alteration of another unidentified gene was adjacent to the ERα gene;30 and 3) intronic changes may have an impact on the expression of receptors by influencing the transcription through alternative splicing of the mRNA transcript.24 Although the susceptibility of Caucasians to prostate cancer is known to be much higher than that of Asians, the underlying mechanism remains unknown.28 Our findings demonstrated the different roles of two ERα SNPs in prostate cancer risk estimation between Caucasians and Asians, indicating the probability of inherited susceptibility to prostate cancer arising from different genomic ERα SNPs, which may help elucidate the pathogenesis of prostate cancer.

Several limitations in this meta-analysis should be noted when interpreting our findings. First, the statistical power was still limited in the stratified analysis. Second, heterogeneity between studies was observed, although subgroup analyses were conducted to minimize the perturbation. Third, only published studies in English were included. Published studies in other languages, ongoing studies, and unpublished data were not obtained. Given these limitations, our conclusions should be interpreted cautiously.

Conclusion

In summary, the current meta-analysis demonstrates the different roles of ERα PvuII and XbaI polymorphisms in prostate cancer risk stratification between Caucasians and Asians, indicating the probability of inherited susceptibility to prostate cancer arising from different genomic ERα SNPs, a finding that may help elucidate the pathogenesis of prostate cancer in Caucasians.

Acknowledgment

This project was supported by the National Natural Science Foundation of China (No 81372335).

Author contributions

Yining Zhao, Xi Zheng, and Xiang Zhang conceived and designed the experiment; Yining Zhao performed the experiment; Lijie Zhang and Hua Jiang screened the studies; Lijie Zhang, Qiang Hu, and Xi Zheng extracted the data; Yining Zhao, Xi Zheng, and Shennan Shi performed the data analysis; and Yining Zhao and Xiang Zhang wrote the paper. All authors contributed toward data analysis, drafting and critically revising the paper and agree to be accountable for all aspects of the work.

Disclosure

The authors report no conflicts of interest in this work.


References

1.

Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.

2.

Qin Z, Li X, Tang J, et al. Association between insulin-like growth factor-binding protein-3 polymorphism-202 A/C and the risk of prostate cancer: a meta-analysis. Onco Targets Ther. 2016;9:5451–5459.

3.

Jia M, Dahlman-Wright K, Gustafsson JA. Estrogen receptor alpha and beta in health and disease. Best Pract Res Clin Endocrinol Metab. 2015;29(4):557–568.

4.

Zhao XZ, Liu Y, Zhou LJ, Wang ZQ, Wu ZH, Yang XY. Role of estrogen in lung cancer based on the estrogen receptor-epithelial mesenchymal transduction signaling pathways. Onco Targets Ther. 2015;8:2849–2863.

5.

Ricke WA, McPherson SJ, Bianco JJ, Cunha GR, Wang Y, Risbridger GP. Prostatic hormonal carcinogenesis is mediated by in situ estrogen production and estrogen receptor alpha signaling. FASEB J. 2008;22(5):1512–1520.

6.

Knobloch K, Yoon U, Vogt PM. Preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement and publication bias. J Craniomaxillofac Surg. 2011;39(2):91–92.

7.

Higgins JPT, Green S. Cochrane Handbook for Systematic Reviews of Interventions, 2011. Available from: http://www.cochrane-handbook.org. Accessed July 1, 2014.

8.

Little J, Higgins JP, Ioannidis JP, et al. STrengthening the REporting of Genetic Association studies (STREGA) – an extension of the STROBE statement. Eur J Clin Invest. 2009;39(4):247–266.

9.

Peters JL, Sutton AJ, Jones DR, Abrams KR, Rushton L. Performance of the trim and fill method in the presence of publication bias and between-study heterogeneity. Stat Med. 2007;26(25):4544–4562.

10.

Modugno F, Weissfeld JL, Trump DL, et al. Allelic variants of aromatase and the androgen and estrogen receptors: toward a multigenic model of prostate cancer risk. Clin Cancer Res. 2001;7(10):3092–3096.

11.

Suzuki K, Nakazato H, Matsui H, et al. Genetic polymorphisms of estrogen receptor alpha, CYP19, catechol-O-methyltransferase are associated with familial prostate carcinoma risk in a Japanese population. Cancer. 2003;98(7):1411–1416.

12.

Tanaka Y, Sasaki M, Kaneuchi M, Shiina H, Igawa M, Dahiya R. Polymorphisms of estrogen receptor alpha in prostate cancer. Mol Carcinog. 2003;37(4):202–208.

13.

Fukatsu T, Hirokawa Y, Araki T, et al. Genetic polymorphisms of hormone-related genes and prostate cancer risk in the Japanese population. Anticancer Res. 2004;24(4):2431–2437.

14.

Hernández J, Balic I, Johnson-Pais TL, et al. Association between an eestrogen receptor alpha gene polymorphism and the risk of prostate cancer in black men. J Urol. 2006;175(2):523–527.

15.

Low YL, Taylor JI, Grace PB, et al. Phytoestrogen exposure, polymorphisms in COMT, CYP19, ESR1, and SHBG genes, and their associations with prostate cancer risk. Nutr Cancer. 2006;56(1):31–39.

16.

Berndt SI, Chatterjee N, Huang WY, et al. Variant in sex hormone-binding globulin gene and the risk of prostate cancer. Cancer Epidemiol Biomarkers Prev. 2007;16(1):165–168.

17.

Kjaergaard AD, Ellervik C, Tybjaerg-Hansen A, et al. Estrogen receptor alpha polymorphism and risk of cardiovascular disease, cancer, and hip fracture: cross-sectional, cohort, and case-control studies and a meta-analysis. Circulation. 2007;115(7):861–871.

18.

Onsory K, Sobti RC, Al-Badran AI, et al. Hormone receptor-related gene polymorphisms and prostate cancer risk in North Indian population. Mol Cell Biochem. 2008;314(1–2):25–35.

19.

Sobti RC, Gupta L, Singh SK, Seth A, Kaur P, Thakur H. Role of hormonal genes and risk of prostate cancer: gene-gene interactions in a North Indian population. Cancer Genet Cytogenet. 2008;185(2):78–85.

20.

Gupta L, Thakur H, Sobti RC, Seth A, Singh SK. Role of genetic polymorphism of estrogen receptor-alpha gene and risk of prostate cancer in north Indian population. Mol Cell Biochem. 2010;335(1–2):255–261.

21.

Balistreri CR, Caruso C, Carruba G, Miceli V, Candore G. Genotyping of sex hormone-related pathways in benign and malignant human prostate tissues: data of a preliminary study. OMICS. 2011;15(6):369–374.

22.

Sissung TM, Danesi R, Kirkland CT, et al. Estrogen receptor alpha and aromatase polymorphisms affect risk, prognosis, and therapeutic outcome in men with castration-resistant prostate cancer treated with docetaxel-based therapy. J Clin Endocrinol Metab. 2011;96(2):E368–E372.

23.

Szendroi A, Speer G, Tabak A, et al. The role of vitamin D, estrogen, calcium sensing receptor genotypes and serum calcium in the pathogenesis of prostate cancer. Can J Urol. 2011;18(3):5710–5716.

24.

Safarinejad MR, Safarinejad S, Shafiei N, Safarinejad S. Estrogen receptors alpha (rs2234693 and rs9340799), and beta (rs4986938 and rs1256049) genes polymorphism in prostate cancer: evidence for association with risk and histopathological tumor characteristics in Iranian men. Mol Carcinog. 2012;51(Suppl 1):E104–E117.

25.

Jurecekova J, Sivonova MK, Evinova A, Kliment J, Dobrota D. The association between estrogen receptor alpha polymorphisms and the risk of prostate cancer in Slovak population. Mol Cell Biochem. 2013;381(1–2):201–207.

26.

Pazarbasi A, Yilmaz MB, Alptekin D, et al. Genetic polymorphisms of estrogen receptor alpha and catechol-O-methyltransferase genes in Turkish patients with familial prostate carcinoma. Indian J Hum Genet. 2013;19(4):408–411.

27.

Lu X, Yamano Y, Takahashi H, et al. Associations between estrogen receptor genetic polymorphisms, smoking status, and prostate cancer risk: a case-control study in Japanese men. Environ Health Prev Med. 2015;20(5):332–337.

28.

Attard G, Parker C, Eeles RA, et al. Prostate cancer. Lancet. 2016;387(10013):70–82.

29.

Fei X, Liu N, Li H, Shen Y, Guo J, Wu Z. Polymorphisms of vitamin D receptor gene TaqI susceptibility of prostate cancer: a meta-analysis. Onco Targets Ther. 2016;9:1033–1045.

30.

Sundermann EE, Maki PM, Bishop JR. A review of estrogen receptor alpha gene (ESR1) polymorphisms, mood, and cognition. Menopause. 2010;17(4):874–886.

Creative Commons License © 2017 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.