Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Genome-Wide Association Study of Breast Cancer in the Japanese Population

  • Siew-Kee Low ,

    amanda.sklow@src.riken.jp

    Affiliations Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Yokohama, Japan, Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan

  • Atsushi Takahashi,

    Affiliation Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Yokohama, Japan

  • Kyota Ashikawa,

    Affiliation Laboratory for Genotyping Development, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Yokohama, Japan

  • Johji Inazawa,

    Affiliation Department of Molecular Cytogenetics, Medical Research Institute and School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan

  • Yoshio Miki,

    Affiliation Department of Genetic iagnosis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan

  • Michiaki Kubo,

    Affiliation Laboratory for Genotyping Development, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Yokohama, Japan

  • Yusuke Nakamura,

    Affiliations Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan, Department of Medicine and Surgery, The University of Chicago, Chicago, Illinois, United States of America

  • Toyomasa Katagiri

    Affiliation Division of Genome Medicine, Institute for Genome Research, The University of Tokushima, Japan

Abstract

Breast cancer is the most common malignancy among women in worldwide including Japan. Several studies have identified common genetic variants to be associated with the risk of breast cancer. Due to the complex linkage disequilibrium structure and various environmental exposures in different populations, it is essential to identify variants associated with breast cancer in each population, which subsequently facilitate the better understanding of mammary carcinogenesis. In this study, we conducted a genome-wide association study (GWAS) as well as whole-genome imputation with 2,642 cases and 2,099 unaffected female controls. We further examined 13 suggestive loci (P<1.0×10−5) using an independent sample set of 2,885 cases and 3,395 controls and successfully validated two previously-reported loci, rs2981578 (combined P-value of 1.31×10−12, OR = 1.23; 95% CI = 1.16–.30) on chromosome 10q26 (FGFR2), rs3803662 (combined P-value of 2.79×10−11, OR = 1.21; 95% CI = 1.15–.28) and rs12922061 (combined P-value of 3.97×10−10, OR = 1.23; 95% CI = 1.15–.31) on chromosome 16q12 (TOX3-LOC643714). Weighted genetic risk score on the basis of three significantly associated variants and two previously reported breast cancer associated loci in East Asian population revealed that individuals who carry the most risk alleles in category 5 have 2.2 times higher risk of developing breast cancer in the Japanese population than those who carry the least risk alleles in reference category 1. Although we could not identify additional loci associated with breast cancer, our study utilized one of the largest sample sizes reported to date, and provided genetic status that represent the Japanese population. Further local and international collaborative study is essential to identify additional genetic variants that could lead to a better, accurate prediction for breast cancer.

Introduction

Breast cancer is the most common malignancy among women worldwide. In Japan, breast cancer comprises approximately 19% of all female cancers; it is the fifth leading cause of cancer death among women with an estimated death of 12,731 in 2011. Its incidence is about 86.0 cases/100,000 individuals/year and 56,289 were newly diagnosed to have breast cancer in 2007 (http://ganjoho.jp/data/public/statistics/backnumber/2012/files/cancer_statistics_2012.pdf). Even though the 5-year survival rate for breast cancer is relatively better compared to other malignancies, the age-adjusted incidence and mortality rate for breast cancer has revealed a significant increase since 1970s in Japan. Hence, breast cancer is one of the most important medical issues to be addressed. In particular, individual risk assessment (genetic and environmental factors), early detection by biomarkers and mammography screening are critically important to reduce breast cancer-associated death.

Although risk factors such as age, age at menarche, ethnicity, reproductive and menstrual history, oral contraceptives, hormone therapy, radiation exposure, mammographic breast density, alcohol intake, dietary folate intake, physical activity and benign breast diseases have been reported [1][7], it is well known that breast cancer is a complex polygenic disease in which genetic factors play an important role in disease etiology and pathogenesis. Individuals who have first-degree relatives with breast cancer are indicated to have approximately 2.1-fold higher risk for the disease [8]. Previous linkage analysis identified mutations in two highly-penetrant genes, BRCA1 and BRCA2, as one of the major cause of inherited cancer in many families [9], [10]. In addition, mutations in ATM, TP53, CHEK2, PTEN, CDH1, STK11 and PALB2 genes also confer risk to breast cancer [11][17]. Nevertheless, mutations in these genes are not common in the general population and account for 5–0% of breast cancer cases. Hence, there is likely to be other genetic variants that contribute to the etiology of this cancer. Since 2007, approximately 67 common genetic variants have been identified to be associated with breast cancer through genome-wide association studies (GWAS) and international collaborative study from European and Asian descendants [18][34]. Due to the complex linkage disequilibrium, differences in allele frequencies and environmental exposure in different populations, it is of importance to identify common genetic variants associated with breast cancer in specific populations, which subsequently facilitate the development of useful prediction systems. Our group has previously reported a GWAS to identify common genetic variants associated with hormonal receptor positive breast cancer [35], but the current study has increased the sample size, which is one of the biggest sample size that represent the Japanese population, and has used a genotyping panel with better coverage aiming to identify common genetic variants associated with all types of breast cancer. Furthermore, we also evaluated the association of previously-identified loci showing the association with breast cancer in the European population and East Asian in the current dataset. Lastly, we conducted whole-genome imputation by referring to 1000G reference panel to increase the coverage of this GWAS study.

Subject and Methods

Study population

We recruited all DNA samples from the Biobank Japan Project (http://biobankjp.org). The Biobank Japan is a bank that has collected DNAs and serum of nearly 200,000 individuals, who had been diagnosed to have one or more of 47 common diseases including various types of cancer from 66 collaborating hospitals in Japan. One of the major objectives of this project is to identify genetic variants that are associated with common diseases and to identify individuals who are at risk for various diseases. In this study, we selected a total of 5,610 breast cancer patients who had been registered in the Biobank Japan. We subsequently divided these patients into two groups of 2,725 and 2,885 cases to be used for the discovery phase and for the validation phase, respectively. For controls, we included 2,331 and 3,395 females consisting of healthy volunteers from Midosuji Rotary Club, Osaka, Japan and Health Science Research Resource Bank as well as individuals in the Biobank who had no history of cancer as controls for discovery and validation phases, respectively. The demographic data of patients recruited for this study is summarized in Table 1. All individuals who participated in this study provided written inform consent. This study was approved by the ethical committees of the Institute of Medical Sciences, the University of Tokyo and RIKEN Center for Integrative Medical Sciences.

thumbnail
Table 1. Demographic data of patients recruited for this study.

https://doi.org/10.1371/journal.pone.0076463.t001

Genotyping and quality control

For the GWAS discovery stage, we genotyped both case and control samples using Illumina OmniExpress BeadChip that contained a total of 733,202 SNPs. After a standard SNP quality control which excluded SNPs with call rate of <0.98, those that deviated from the Hardy-Weinberg equilibrium (P≤1.0×10−6), those on the X chromosome and non-polymorphic SNPs, a total of 550,026 SNPs were used for further analysis. The cluster plot of 100 SNPs that revealed the strongest associations were checked by visual observation to exclude SNPs with ambiguous genotypes. For sample quality control, we evaluated cryptic relatedness for each sample with identity-by-state method. To examine population stratification of this study, we performed principal component analysis (PCA) using EIGENSTRAT software v2.0 (http://genepath.med.harvard.edu/~reich/Software.htm) with four reference populations from the HapMap data as reference including Europeans (represented by Caucasian from UTAH, CEU), Africans (represented by Yoruba from Ibadan, YRI) and East Asians (represented by Japanese from Tokyo, JPT, and Han Chinese from Beijing, CHB) (Figure S1a). We plotted the scatter plot by using the top two associated principal components (eigenvectors) to identify outliers who did not belong to the JPT/CHB cluster. Subsequently, we performed PCA analysis using only the genotype information of the case and control subjects to further evaluate the population substructure (Figure S1b). Quantile-quantile (Q-Q) plot was constructed using observed P-values against expected P-values and an inflation factor value (λ-value) that was calculated to assess potential population stratification of the study subjects (Figure S2a and S2b). After performing PCA, we selected 2,642 cases and 2,099 controls within the major Japanese (Hondo) cluster for subsequent analysis (Figure S1b).

Imputation analysis

To increase the power and coverage of the genome-wide association scan, we performed whole genome imputation using 1000G of East Asian population (Japanese in Tokyo JPT, Chinese in Beijing CHB and Chinese in Denver CHD) Phase I Integrated Release Version 2 dataset as reference panel to infer missing genotypes. Briefly, we prepared the input files after quality control, which excluded SNPs with genotyping rate of <98%, those that deviated from HWE (HWE P≤1.0×10−6) and those with MAF of <0.01. We then confirmed that the allele frequencies of the reference allele are comparable between the GWAS dataset and the reference panel with differences of <0.15. By using MACH1.0 (http://www.sph.umich.edu/csg/abecasis/MACH/index.html), we performed haplotype phasing with the samples' genotypes referring 1000G reference panel, estimated the map crossover and error rates using 20 iterations of the Markov chain. Subsequently, we imputed the missing genotypes using Minimac (http://genome.sph.umich.edu/wiki/Minimac). We utilized stringent imputation quality control by excluding SNPs with r2 value of <0.9.

Validation study

After evaluating the associations from GWAS and whole genome imputation, we selected a total of 13 candidate loci that showed suggestive association (P<1.0×10−5) with breast cancer risk for further validation by an independent set of 2,885 cases and 3,395 controls. We genotyped the cases with the multiplex-PCR Invader assay [36] and the control samples with either Illumina OmniExpress BeadChip Kits or by imputation. To verify the accuracy of the imputation analysis, we also included surrogate SNPs that showed close link (r2>0.8 and D' = 1.00) to the imputed SNPs and were included in the genotype platform. Considering multiple testing at this validation stage, we applied Bonferroni significance threshold at P<3.85×10−3 (0.05/13 independent tests).

Evaluation of previously reported loci

To verify previously-reported loci showing the association with breast cancer in the European and East Asian populations, we evaluated 67 loci in the current Japanese GWAS dataset [18][34]. Among the 67 SNPs examined, 6 SNPs are not polymorphic in the Japanese population, 26 SNPs are same as the previously-reported SNPs, 33 and 2 SNPs are SNPs having r2-value of more than 0.8 and 0.7 to the previously-reported SNPs, respectively (Table S4).

Statistical Analysis

The case-control associations of the GWAS discovery set and validation set were evaluated using logistic regression analysis after considering age as confounding factor from PLINK software (http://pngu.mgh.harvard.edu/~purcell/plink/). The associations of the imputed SNPs were generated with mach2dat software which utilized the output results from Minimac (dosage of the imputed SNP). To have an overview of the association of SNPs with breast cancer, a Manhattan plot of the study was plotted using Haploview 4.1. Meta-analysis for the combined analysis of the discovery and validation phase was performed using inverse-variance method and heterogeneity between the two phases was evaluated using Cochran's Q test. Regional association plots were generated using Locus Zoom (http://csg.sph.umich.edu/locuszoom/).

Weighted genetic risk score (wGRS)

To evaluate the cumulative effects of genetic variants associated with breast cancer risk, we conducted weighted genetic risk score (wGRS) analysis on the basis of genotypes of five SNPs, three significant SNPs (rs2981578 of 10q26/FGFR2, rs3803662 and rs12922061 of 16q12/TOX3) from this study and two SNPs (rs6557161 of 6q25/ESR1 and rs10509168 of 10q21/ZNF365) that were reported to be associated with breast cancer risk in East Asian population and indicated suggestive association in this study. The wGRS model was developed by logistic regression analysis by incorporating five associated-SNPs and age (as covariates) using GWAS dataset to obtain the estimates (weight) of each corresponding SNP. This model was subsequently validated in an independent samples dataset drawn from the validation phase of this study. The cumulative genetic risk scores were determined by multiplying the number of risk alleles (0/1/2) of an individual by its corresponding weight, and subsequently the sum across the total number of SNPs were taken into consideration. We then classified the genetic risk score into five different categories created from the mean and standard deviation (SD); group 1, < mean-1SD; group 2, mean-1SD to mean; group 3, mean to mean+1SD; group 4, mean+1SD to mean+2SD, group 5, > mean+2SD. Odds ratio and 95% confidence interval were calculated using group 1 as a reference.

Results

In this study, we genotyped a total of 2,725 cases and 2,311 controls with Illumina OmniExpress BeadChip Kits that contained 733,202 SNPs to identify genetic variants associated with the susceptibility to breast cancer in the Japanese population. After quality check of the SNP genotyping data, a total of 550,026 autosomal SNPs were examined for the association by logistic regression analysis. Quantile-quantile (Q-Q) plot and the genomic inflation factor (λ) of the test statistic of this GWAS based on 550, 026 SNPs with all samples was 1.183 suggesting the existence of some population substructure (Figure S2a). To exclude the possibility of population substructure for our sample population, we performed principal component analysis (PCA). Although all the subjects participating in this study were clustered in the Asian population, there was a small portion of samples that were separated from the major Japanese (Hondo) cluster when PCA analysis was performed using only the genotype information of the case and control in the study (Figure S1a and S1b). We then used samples from the major Japanese (Hondo) cluster consisting of 2,642 cases and 2,099 controls, and found that the λ-value improved to 1.027 (Figure S2b). Hence, subsequent analysis was carried out using only samples from the major Japanese cluster. Whole genome imputation utilizing 1000G database as reference panel successfully estimated 7,791,127 SNPs. After stringent quality control by excluding SNPs with r2-value of <0.9, the total number of SNPs that were taken into account was 5,335,291. The Manhattan plot, plotting –log10 (P-value) from the GWAS and imputation analysis against the chromosome position, showed that there were no genetic loci achieving genome-wide significance with the threshold P-value of <5×10−8 (Figure 1).

thumbnail
Figure 1. The Manhattan plot for GWAS of breast cancer in the Japanese population.

This plot is based on –log10 (P-value) from GWAS and imputation analysis against chromosome position, each color represents different chromosome. Blue line indicate suggestive association threshold, P = 1×10−5 while red line indicate genome-wide significant threshold P<5×10−8.

https://doi.org/10.1371/journal.pone.0076463.g001

To identify additional susceptible loci associated with breast cancer, we conducted a validation study of 13 genetic loci showing suggestive association (P<1×10−5) with breast cancer after excluding SNPs that showed linkage disequilibrium (LD) coefficient (r2) of >0.8 within each LD block by examining an independent set of 2,885 breast cancer cases and 3,395 controls. Among the 13 loci tested, three SNPs (rs2981578 on chromosome 10q26.13 and rs3803662 along with rs12922061 on chromosome 16q12.1) were successfully validated with Bonferroni-corrected P-value of <3.85×10−3 (0.05/13 independent tests) as shown in Table 2 and Table S1. Inverse variance meta-analysis indicated that these three SNPs surpass genome-wide significance level (P-value<5×10−8) after combining the GWAS and the validation study with no significant heterogeneity (P-value >0.05) between the two stages (Table 2).

thumbnail
Table 2. Association study of SNPs on chromosome 10q26.13 and 16q12.1.

https://doi.org/10.1371/journal.pone.0076463.t002

The most significantly associated SNP, rs2981578 (combined P-value of 1.31×10−12, OR = 1.23; 95% CI = 1.16–.30), is located within the second intron of the FGFR2 gene on chromosome 10q26.13 (Table 2 and Figure 2a). Variants on this gene have been the most frequently validated to be associated with breast cancer in multiple populations. For chromosome 16q21.1, we successfully validated rs12922061 (combined P-value of 3.97×10−10, OR = 1.23; 95% CI = 1.15–.31) to be significantly associated with breast cancer (Table 2 and Figure 2b). After conditioning the effect of rs12922061, rs3803662 remained suggestively associated and was successfully validated after additional samples with a combined P-value of 2.79×10−11 (OR = 1.21; 95% CI = 1.14–.25). Two of these SNPs remained significant (P-value<0.0001) after performing condition analysis by using one of the SNP as covariate, suggesting the independency of association with breast cancer (Table S2). Additionally, the r2 value between these two SNPs is only 0.17, indicating they are not closely linked with each other. Haplotype analysis of the two SNPs did not reveal stronger association than a single SNP association after 100,000 permutation analysis (Table S3). The SNP, rs3803662, is located in the last exon of LOC643714 and near to the 5′ end of TOX3; whilst rs12922061 is located in the first intron of LOC643714.

thumbnail
Figure 2. Regional association plots for two significantly associated loci with breast cancer in Japanese population, (a) chromosome 10q26.13 (FGFR2) and (b) chromosome 16q21.1 (TOX3-LOC643714).

SNPs from the GWAS are plotted as circles; imputed SNPs are plotted as crosses. The color intensity reflects the extent of LD with the marker SNP: red, (r2≥ 0.8), orange (0.6≤r2≤0.8), green (0.4≤r2≤0.6), light blue (0.2≤r2≤0.4) and dark blue (r2<0.2). Purplish blue lines represent local recombination rates. The SNP position is based on NCBI build 37.

https://doi.org/10.1371/journal.pone.0076463.g002

In addition to perform GWAS for breast cancer in Japanese population, we also evaluated the association of previously-reported breast cancer risk loci in the European and East Asian populations. We evaluated a total of 61 SNPs after excluding 6 SNPs that are not polymorphic in Japanese population (Table S4). Among the 61 SNPs, eight SNPs (rs4415084 of 5p12/MRPS30, rs6557161 of 6q25/ESR1, rs7465364 of 8p21/RPL17p33, rs672888 of 8q24/MYC, rs10509168 of 10q21/ZNF365, rs1219648 of 10q26/FGFR2, rs17221259 of 12p13/ATF7IP and rs3803662 of 16q12/TOX3) showed suggestive association (P-value<0.05) with breast cancer in Japanese population (Table 3). All of these suggestively-associated SNPs possessed the same risk allele and showed the same direction of association that was indicated in the previous reports.

thumbnail
Table 3. Association of previously reported to be breast cancer susceptibility loci in current Japanese GWAS dataset.

https://doi.org/10.1371/journal.pone.0076463.t003

After developing wGRS model using five SNPs from the GWAS dataset, the model was subsequently validated in an independent sample set represented by the validation samples. The cumulative effect of five SNPs evaluated by the wGRS analysis indicated that odds ratio of each category increased according to the level of risk score, and individuals who are in category five carrying the most risk alleles have 2.2 times higher risk to develop breast cancer when utilizing category 1 as a reference (Table 4).

thumbnail
Table 4. wGRS using 5 significant associated SNPs evaluated on independent validation sample set.

https://doi.org/10.1371/journal.pone.0076463.t004

Discussion

To investigate the involvement of common genetic variants (SNPs) associated with breast cancer in the Japanese population, we performed GWAS, whole genome imputation using 1000G database as reference panel and validation study using a total of 5,527 breast cancer cases and 5,494 controls individuals. We successfully validated the association of chromosome 10q26.13 (FGFR2), and 16q12.1 (TOX3-LOC643714). In addition to the two aforementioned loci, we validated a total of 67 loci that were previously reported the association with breast cancer and identified six additional loci (rs4415084 of 5p12/MRPS30, rs6557161 of 6q25/ESR1, rs7465364 of 8p21/RPL17p33, rs672888 of 8q24/MYC, rs10509168 of 10q21/ZNF365 and rs17221259 of 12p13/ATF7IP) to have suggestive association (P<0.05) with breast cancer in Japanese population. Further fine mapping of these loci might identify insightful findings for future analysis.

Hunter DJ et al. first reported the association of FGFR2 with breast cancer in 2007 [19]. Since then, this locus has been successfully validated in various populations throughout the world including those of European ancestry, Asian, Ashkenazi Jewish and Israeli populations [18], [37][39]. FGFR2 encodes fibroblast growth factor receptor type 2, which is a receptor tyrosine kinase playing a critical role in the growth signaling pathway that is involved in growth and differentiation of cells in various tissues including the breast and kidney [40], [41]. All the SNPs that were found to be associated with breast cancer are located in intron 2 of the gene; the risk allele of rs2981578, a SNP that was identified in this study, created a putative binding site for Oct-1/Runx2, which gives rise to a strong protein-DNA complex that alters binding of the transcription factor and causes differential expression between the common and minor haplotypes of FGFR2 [42]. Additionally, Zhu et al. also reported that there is a potential role of histone 3/4 acetylation in modulating access to the polymorphic sites within intron 2 in addition to downstream splicing sites in generating variable FGFR2 levels and isoforms in breast cancer [43].

The second significantly associated locus is located on chromosome 16q12.1 (TOX3-LOC643714). LOC643714 is an uncharacterized gene of unknown function; TOX3, also known as TNRC9 or CAGF9, encodes a high mobility group box nuclear protein, which is involved in regulating calcium-dependent transcription [44]. A previous study indicated that increased expression of TOX3 could be a predictor of breast cancer metastasis to bone [45]. In this study, we identified two independently associated SNPs, rs3803662 and rs12922061, with breast cancer in the Japanese population. The minor allele of rs3803662 is reported to cause lower mRNA expression of TOX3 gene, and this regulatory SNP may alter the expression of a distant gene, RBL2, in cis [46].

Although wGRS of five associated loci with breast cancer in the Japanese population revealed that individuals with the highest risk (category 5) have 2.2 times higher risk than those with the lowest risk (category 1), it is believed that a complex disease such as breast cancer would be affected by a large number of common genetic variants that have very modest effects. This phenomenon was also supported by the six additional reported loci that showed suggestive association in this dataset, indicating that our current dataset is still under statistical power. Hence, to increase the power and to enlarge the sample number, there is a need for more local and international institutions to collaborate with each other in identifying more common variants associated with breast cancer, which hopefully will lead to the development of promising and accurate prediction system.

Supporting Information

Figure S1.

Principal component analysis of (a) Case and control samples of this study with four reference populations from the HapMap database which include Europeans (represented by Caucasian from UTAH, CEU), Africans (represented by Yoruba from Ibadan, YRI) and East Asians (represented by Japanese from Tokyo, JPT, and Han Chinese from Beijing, CHB). (b) Case and control samples of this study. Samples from the major cluster (within the black oval circle) were selected for further analysis.

https://doi.org/10.1371/journal.pone.0076463.s001

(TIFF)

Figure S2.

Quantile-quantile (Q-Q) plot for GWAS of breast cancer in Japanese population with (a) All samples (λ = 1.18) and (b) Major Japanese (Hondo) cluster (λ = 1.03).

https://doi.org/10.1371/journal.pone.0076463.s002

(TIFF)

Table S1.

Association study of the 13 selected loci.

https://doi.org/10.1371/journal.pone.0076463.s003

(XLS)

Table S2.

Conditioning analysis of SNPs on chromosome 16q12.1.

https://doi.org/10.1371/journal.pone.0076463.s004

(XLS)

Table S3.

Haplotype analysis and association of SNPs on chromosome 16q12.1.

https://doi.org/10.1371/journal.pone.0076463.s005

(XLS)

Table S4.

Association study of previously reported breast cancer associated loci.

https://doi.org/10.1371/journal.pone.0076463.s006

(XLS)

Acknowledgments

We express our heartfelt gratitude to all the patients who participate in this study. We also thank the members of The Rotary Club of Osaka-Midosuji District 2660 Rotary International in Japan for making this study possible. We would like to express our gratefulness to the staff from Biobank Japan for their outstanding assistance. We extend our appreciation to Mr. Kohei Tomizuka for his insightful comment and members from the Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN for their technical support.

Author Contributions

Conceived and designed the experiments: YN TK. Performed the experiments: SKL KA. Analyzed the data: SKL AT. Contributed reagents/materials/analysis tools: AT JI YM MK. Wrote the paper: SKL YN TK.

References

  1. 1. Phipps AI, Chlebowski RT, Prentice R, McTiernan A, Wactawski-Wende J, et al. (2011) Reproductive history and oral contraceptive use in relation to risk of triple-negative breast cancer. J Natl Cancer Inst 103: 470–77.
  2. 2. Iodice S, Barile M, Rotmensz N, Feroce I, Bonanni B, et al. (2010) Oral contraceptive use and breast or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Cancer 46: 2275–284.
  3. 3. Anderson GL, Limacher M, Assaf AR, Bassford T, Beresford SA, et al. (2004) Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 291: 1701–712.
  4. 4. Hamajima N, Hirose K, Tajima K, Rohan T, Calle EE, et al. (2002) Alcohol, tobacco and breast cancer – llaborative reanalysis of individual data from 53 epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the disease. Br J Cancer 87: 1234–245.
  5. 5. McTiernan A (2003) Behavioral risk factors in breast cancer: can risk be modified? Oncologist 8: 326–334.
  6. 6. Islam T, Ito H, Sueta A, Hosono S, Hirose K, et al.. (2012) Alcohol and dietary folate intake and the risk of breast cancer: a case-control study in Japan. Eur J Cancer Prev.
  7. 7. Chen J, Pee D, Ayyagari R, Graubard B, Schairer C, et al. (2006) Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst 98: 1215–1226.
  8. 8. Pharoah PD, Day NE, Duffy S, Easton DF, Ponder BA (1997) Family history and the risk of breast cancer: a systematic review and meta-analysis. Int J Cancer 71: 800–809.
  9. 9. Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K, et al. (1994) A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 266: 66–71.
  10. 10. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, et al. (1995) Identification of the breast cancer susceptibility gene BRCA2. Nature 378: 789–792.
  11. 11. Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, et al. (2006) ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet 38: 873–875.
  12. 12. Coles C, Condie A, Chetty U, Steel CM, Evans HJ, et al. (1992) p53 mutations in breast cancer. Cancer Res 52: 5291–5298.
  13. 13. Consortium CBCC-C (2004) CHEK2*1100delC and susceptibility to breast cancer: a collaborative analysis involving 10,860 breast cancer cases and 9,065 controls from 10 studies. Am J Hum Genet 74: 1175–1182.
  14. 14. Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, et al. (1997) Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 16: 64–67.
  15. 15. Berx G, Cleton-Jansen AM, Nollet F, de Leeuw WJ, van de Vijver M, et al. (1995) E-cadherin is a tumour/invasion suppressor gene mutated in human lobular breast cancers. EMBO J 14: 6107–6115.
  16. 16. Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, et al. (2006) Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res 12: 3209–3215.
  17. 17. Hofstatter EW, Domchek SM, Miron A, Garber J, Wang M, et al. (2011) PALB2 mutations in familial breast and pancreatic cancer. Fam Cancer 10: 225–231.
  18. 18. Easton DF, Pooley KA, Dunning AM, Pharoah PD, Thompson D, et al. (2007) Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447: 1087–1093.
  19. 19. Hunter DJ, Kraft P, Jacobs KB, Cox DG, Yeager M, et al. (2007) A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet 39: 870–874.
  20. 20. Stacey SN, Manolescu A, Sulem P, Rafnar T, Gudmundsson J, et al. (2007) Common variants on chromosomes 2q35 and 16q12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 39: 865–869.
  21. 21. Stacey SN, Manolescu A, Sulem P, Thorlacius S, Gudjonsson SA, et al. (2008) Common variants on chromosome 5p12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 40: 703–706.
  22. 22. Zheng W, Long J, Gao YT, Li C, Zheng Y, et al. (2009) Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat Genet 41: 324–328.
  23. 23. Thomas G, Jacobs KB, Kraft P, Yeager M, Wacholder S, et al. (2009) A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11.2 and 14q24.1 (RAD51L1). Nat Genet 41: 579–584.
  24. 24. Ahmed S, Thomas G, Ghoussaini M, Healey CS, Humphreys MK, et al. (2009) Newly discovered breast cancer susceptibility loci on 3p24 and 17q23.2. Nat Genet 41: 585–590.
  25. 25. Turnbull C, Ahmed S, Morrison J, Pernet D, Renwick A, et al. (2010) Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet 42: 504–507.
  26. 26. Long J, Cai Q, Shu XO, Qu S, Li C, et al. (2010) Identification of a functional genetic variant at 16q12.1 for breast cancer risk: results from the Asia Breast Cancer Consortium. PLoS Genet 6: e1001002.
  27. 27. Fletcher O, Johnson N, Orr N, Hosking FJ, Gibson LJ, et al. (2011) Novel breast cancer susceptibility locus at 9q31.2: results of a genome-wide association study. J Natl Cancer Inst 103: 425–435.
  28. 28. Cai Q, Long J, Lu W, Qu S, Wen W, et al. (2011) Genome-wide association study identifies breast cancer risk variant at 10q21.2: results from the Asia Breast Cancer Consortium. Hum Mol Genet 20: 4991–4999.
  29. 29. Long J, Cai Q, Sung H, Shi J, Zhang B, et al. (2012) Genome-wide association study in east Asians identifies novel susceptibility loci for breast cancer. PLoS Genet 8: e1002532.
  30. 30. Ghoussaini M, Fletcher O, Michailidou K, Turnbull C, Schmidt MK, et al. (2012) Genome-wide association analysis identifies three new breast cancer susceptibility loci. Nat Genet 44: 312–318.
  31. 31. Lindstrom S, Vachon CM, Li J, Varghese J, Thompson D, et al. (2011) Common variants in ZNF365 are associated with both mammographic density and breast cancer risk. Nat Genet 43: 185–187.
  32. 32. Michailidou K, Hall P, Gonzalez-Neira A, Ghoussaini M, Dennis J, et al.. (2013) Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat Genet 45: 353–361, 361e351–352.
  33. 33. Li J, Humphreys K, Heikkinen T, Aittomaki K, Blomqvist C, et al. (2011) A combined analysis of genome-wide association studies in breast cancer. Breast Cancer Res Treat 126: 717–727.
  34. 34. Haiman CA, Chen GK, Vachon CM, Canzian F, Dunning A, et al. (2011) A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor-negative breast cancer. Nat Genet 43: 1210–1214.
  35. 35. Elgazzar S, Zembutsu H, Takahashi A, Kubo M, Aki F, et al. (2012) A genome-wide association study identifies a genetic variant in the SIAH2 locus associated with hormonal receptor-positive breast cancer in Japanese. J Hum Genet 57: 766–771.
  36. 36. Ohnishi Y, Tanaka T, Ozaki K, Yamada R, Suzuki H, et al. (2001) A high-throughput SNP typing system for genome-wide association studies. J Hum Genet 46: 471–477.
  37. 37. Liang J, Chen P, Hu Z, Zhou X, Chen L, et al. (2008) Genetic variants in fibroblast growth factor receptor 2 (FGFR2) contribute to susceptibility of breast cancer in Chinese women. Carcinogenesis 29: 2341–2346.
  38. 38. Gold B, Kirchhoff T, Stefanov S, Lautenberger J, Viale A, et al. (2008) Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33. Proc Natl Acad Sci U S A 105: 4340–4345.
  39. 39. Raskin L, Pinchev M, Arad C, Lejbkowicz F, Tamir A, et al. (2008) FGFR2 is a breast cancer susceptibility gene in Jewish and Arab Israeli populations. Cancer Epidemiol Biomarkers Prev 17: 1060–1065.
  40. 40. Bates CM (2007) Role of fibroblast growth factor receptor signaling in kidney development. Pediatr Nephrol 22: 343–349.
  41. 41. Parsa S, Ramasamy SK, De Langhe S, Gupte VV, Haigh JJ, et al. (2008) Terminal end bud maintenance in mammary gland is dependent upon FGFR2b signaling. Dev Biol 317: 121–131.
  42. 42. Meyer KB, Maia AT, O'Reilly M, Teschendorff AE, Chin SF, et al. (2008) Allele-specific up-regulation of FGFR2 increases susceptibility to breast cancer. PLoS Biol 6: e108.
  43. 43. Zhu X, Asa SL, Ezzat S (2009) Histone-acetylated control of fibroblast growth factor receptor 2 intron 2 polymorphisms and isoform splicing in breast cancer. Mol Endocrinol 23: 1397–1405.
  44. 44. Yuan SH, Qiu Z, Ghosh A (2009) TOX3 regulates calcium-dependent transcription in neurons. Proc Natl Acad Sci U S A 106: 2909–2914.
  45. 45. Smid M, Wang Y, Klijn JG, Sieuwerts AM, Zhang Y, et al. (2006) Genes associated with breast cancer metastatic to bone. J Clin Oncol 24: 2261–2267.
  46. 46. Gudmundsdottir ET, Barkardottir RB, Arason A, Gunnarsson H, Amundadottir LT, et al. (2012) The risk allele of SNP rs3803662 and the mRNA level of its closest genes TOX3 and LOC643714 predict adverse outcome for breast cancer patients. BMC Cancer 12: 621.