In English | En español
Questions About Cancer? 1-800-4-CANCER

Genetics of Breast and Ovarian Cancer (PDQ®)

  • Last Modified: 08/08/2012

Page Options

  • Print This Page
  • Print This Document
  • View Entire Document
  • Email This Document

Low-Penetrance Predisposition to Breast and Ovarian Cancer

Background
Breast Cancer Susceptibility Genes Identified Through Candidate Gene Approaches
        CHEK2
        ATM
        BRIP1
        PALB2
        CASP8 and TGFB1
        RAD51C
Genome-Wide Searches



Background

Mutations in BRCA1, BRCA2, and the genes involved in other rare syndromes discussed in the Major Genes section of this summary account for less than 25% of the familial risk of breast cancer.[1] Despite intensive genetic linkage studies, there do not appear to be other BRCA1/BRCA2-like high-penetrance genes that account for a significant fraction of the remaining multiple-case familial clusters.[2] These observations suggest that the remaining breast cancer susceptibility is polygenic in nature, meaning that a relatively large number of low-penetrance genes are involved.[3] On its own, each low-penetrance locus would be expected to have a relatively small effect on breast cancer risk and would not produce dramatic familial aggregation or influence patient management. However in combination with other genetic loci and/or environmental factors, particularly given how common these can be, variants of this kind might significantly alter breast cancer risk. These types of genetic variations are sometimes referred to as “polymorphisms,” meaning that the gene or locus occurs in several “forms” within the population (and more formally defined as polymorphic when a specific variation in a given locus occurs in more than 1% of the population). Most loci that are polymorphic have no influence on disease risk or human traits (benign polymorphisms), while those that are associated with a difference in risk of disease or a human trait (however subtle) are sometimes termed “disease-associated polymorphisms” or “functionally relevant polymorphisms.” This polygenic model of susceptibility is consistent with the observed patterns of familial aggregation of breast cancer.[4] Although the clinical significance and causality of associations with breast cancer are often difficult to evaluate and establish, genetic polymorphisms may account for why some individuals are more sensitive than others to environmental carcinogens.[5]

Polymorphisms underlying polygenic susceptibility to breast cancer are considered low penetrance, a term often applied to sequence variants associated with a minimal to moderate risk. This is in contrast to “high-penetrance” variants or alleles that are typically associated with more severe phenotypes, for example those BRCA1/BRCA2 mutations leading to an autosomal dominant inheritance patterns in a family. The definition of a “moderate” risk of cancer is arbitrary, but it is usually considered to be in the range of a relative risk of 1.5 to 2.0. Because these types of sequence variants (also called low-penetrance genes, alleles, mutations, and polymorphisms) are relatively common in the general population, their contribution to cancer risk overall is estimated to be much greater than the attributable risk in the population from mutations in BRCA1 and BRCA2. For example, it is estimated by segregation analysis that half of all breast cancer occurs in 12% of the population that is deemed most susceptible.[3] There are no known low-penetrance variants in BRCA1/BRCA2. The N372H variation in BRCA2, initially thought to be a low-penetrance allele, was not verified in a large combined analysis.[6]

Two strategies have been taken to identify low-penetrance polymorphisms leading to breast cancer susceptibility: candidate gene and genome-wide searches. Both involve the epidemiologic case-control study design. The candidate gene approach involves selecting genes based on their known or presumed biological function, relevance to carcinogenesis or organ physiology, and searching for or testing known genetic variants for an association with cancer risk. This strategy relies on imperfect and incomplete biological knowledge, and, despite some confirmed associations (described below), has been relatively disappointing [6,7] The candidate gene approach has largely been replaced by the genome-wide association studies (GWAS) in which a very large number of single nucleotide polymorphisms (SNPs) (potentially 1 million or more) are chosen within the genome and tested, mostly without regard to their possible biological function, but instead to capture all genetic variation throughout the genome more uniformly.

Breast Cancer Susceptibility Genes Identified Through Candidate Gene Approaches

There is a very large literature of genetic epidemiology studies describing associations between various loci and breast cancer risk. Many of these studies suffer from significant design limitations. Perhaps as a consequence, most reported associations do not replicate in follow-up studies. This section is not a comprehensive review of all reported associations. This section describes associations that are believed by the editors to be clinically valid, in that they have been described in several different studies or are supported by robust meta-analyses. The clinical utility of these observations remains unclear, however, as the risks associated with these variations usually fall below a threshold that would justify a clinical response.

CHEK2

CHEK2 (OMIM) is a gene involved in the DNA damage repair response pathway. Based on numerous studies, a polymorphism, 1100delC, appears to be a rare, moderate-penetrance cancer susceptibility allele.[8-13] One study identified the mutation in 1.2% of the European controls, 4.2% of the European BRCA1/BRCA2-negative familial breast cancer cases, and 1.4% of unselected female breast cancer cases.[8] In a group of 1,479 Dutch women younger than 50 years with invasive breast cancer, 3.7% were found to have the CHEK2 1100delC mutation.[14] In additional European and U.S. (where the mutation appears to be slightly less common) studies, including a large prospective study,[15] the frequency of CHEK2 mutations detected in familial breast or ovarian cancer cases has ranged from 0% [16] to 11%; overall, these studies have found an approximately 1.5-fold to 3-fold increased risk of female breast cancer.[15,17-20] A multicenter combined analysis and reanalysis of nearly 20,000 subjects from ten case-control studies, however, has verified a significant 2.3-fold excess of breast cancer among mutation carriers.[21]

Two studies have suggested that the risk associated with a CHEK2 1100delC mutation was stronger in the families of probands ascertained because of bilateral breast cancer.[22,23] Furthermore, a meta-analysis of 1100delC mutation carriers estimated the risk of breast cancer to be 42% by age 70 years in women with a family history of breast cancer.[24] Similarly, a Polish study reported that CHEK2 truncating mutations confer breast cancer risks based on a family history of breast cancer as follows: no family history: 20%; one second-degree relative: 28%; one first-degree relative: 34%; and both first- and second-degree relatives: 44%.[25] Although there have been conflicting reports regarding cancers other than breast cancer associated with CHEK2 mutations, this may be dependent on mutation type (i.e., missense vs. truncating) or population studied and is not currently of clinical utility.[13,18,26-31] The contribution of CHEK2 mutations to breast cancer may depend on the population studied, with a potentially higher mutation prevalence in Poland.[32] CHEK2 mutation carriers in Poland may be more susceptible to ER-positive breast cancer.[33]

Currently, the clinical applicability of CHEK mutations remains uncertain because of low mutation prevalence and lack of guidelines for clinical management.[34]

ATM

Ataxia telangiectasia (AT) (OMIM) is an autosomal recessive disorder characterized by neurologic deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. It is estimated that 1% of the general population may be heterozygote carriers of ATM mutations (OMIM).[35] More than 300 mutations in the gene have been identified to date, most of which are truncating mutations.[36] ATM proteins have been shown to play a role in cell cycle control.[37-39] In vitro, AT-deficient cells are sensitive to ionizing radiation and radiomimetic drugs, and lack cell cycle regulatory properties after exposure to radiation.[40]

Initial studies searching for an excess of ATM mutations among breast cancer patients provided conflicting results, perhaps due to study design and mutation testing strategies.[41-51] However, two large epidemiologic studies have demonstrated a statistically increased risk of breast cancer among female heterozygote carriers, with an estimated relative risk of approximately 2.0.[51,52] Despite this convincing epidemiologic association, the clinical application of testing for ATM mutations is unclear due to the wide mutational spectrum and the logistics of testing. Because the presence of a mutation could pose a risk in screening-related radiation exposure, further investigation is needed.

BRIP1

BRIP1 (also known as BACH1) encodes a helicase that interacts with the BRCT domain of BRCA1. This gene also has a role in BRCA1-dependent DNA repair and cell cycle checkpoint function. Biallelic mutations in BRIP1 are a cause of Fanconi anemia,[53-55] much like such mutations in BRCA2. Inactivating mutations of BRIP1 are associated with an increased risk of breast cancer. In one study, more than 3,000 individuals from BRCA1/BRCA2 mutation negative families were examined for BRIP1 mutations. Mutations were identified in 9 of 1,212 individuals with breast cancer but in only 2 of 2,081 controls (P = .003). The relative risk of breast cancer was estimated to be 2.0 (95% confidence interval [CI], 1.2–3.2; P = .012). Of note, in families with BRIP1 mutations and multiple cases of breast cancer, there was incomplete segregation of the mutation with breast cancer, consistent with a low penetrance allele and similar to that seen with CHEK2.[56]

PALB2

PALB2 (partner and localizer of BRCA2) interacts with the BRCA2 protein and plays a role in homologous recombination and double-stranded DNA repair. Similar to BRIP1 and BRCA2, biallelic mutations in PALB2 have also been shown to cause Fanconi anemia.[57] PALB2 mutations have been screened for in multiple small studies of familial and early-onset breast cancer in multiple populations.[58-68] Mutation prevalence has ranged from 0.4% to 3.4%. Similar to BRIP1 and CHEK2, there was incomplete segregation of PALB2 mutations in families with hereditary breast cancer.[59] A Finnish PALB2 founder mutation (c.1592delT) has been reported to confer a 40% risk of breast cancer to age 70 years [60] and is associated with a high incidence (54%) of triple-negative disease and lower survival.[61] Mutations have been observed in early-onset and familial breast cancer in many populations.[62,63]

Male breast cancer has been observed in PALB2 mutation–positive breast cancer families.[58,64] In a study of 115 male breast cancer cases in which 18 men had BRCA2 mutations, an additional two men had either a pathogenic or predicted pathogenic PALB2 mutation (accounting for about 10% of germline mutations in the study and 1%–2% of the total sample).[58] Following the identification of PALB2 mutations in pancreatic tumors and the detection of germline mutations in 3% of 96 familial pancreatic patients,[69] numerous studies have pointed to a role for PALB2 in pancreatic cancer. A sixfold increase in pancreatic cancer was observed in the relatives of 33 BRCA1/2-negative, PALB2 mutation–positive breast cancer probands.[64] PALB2 mutations were detected in 3.7% of 81 familial pancreatic cancer families [70] and in 2.1% of 94 BRCA1/2 mutation–negative breast cancer patients who had either a personal or family history of pancreatic cancer.[71] Two relatively small studies, one of 77 BRCA1/2 mutation–negative probands with a personal or family history of pancreatic cancer, one-half of whom were of Ashkenazi Jewish descent, and another study of 29 Italian pancreatic cancer patients with a personal or family history of breast or ovarian cancer, failed to detect any PALB2 mutations.[72,73]

The observed prevalence of PALB2 mutations in familial breast cancer varied depending on ascertainment relative to personal and family history of pancreatic and ovarian cancers, but in all studies, the observed mutation rate was less than 4%. The relative risk of breast cancer appears moderate, and the risk of other cancers (e.g., pancreatic) is poorly defined; therefore, the clinical utility of testing is not clear. There is insufficient evidence to support routine screening of PALB2 when tests of the more common genes, namely BRCA1/2, are negative.

CASP8 and TGFB1

The Breast Cancer Association Consortium (BCAC) investigated single nucleotide polymorphisms identified in previous studies as possibly associated with excess breast cancer risk in 15,000 to 20,000 cases and 15,000 to 20,000 controls. Two SNPs, CASP8 D302H and TGFB1 L10P, were associated with invasive breast cancer with relative risks of 0.88 (95% CI, 0.84–0.92) and 1.08 (95% CI, 1.04–1.11) respectively.[74]

RAD51C

RAD51C is involved in DNA damage repair through homologous recombination and interaction with numerous DNA repair proteins. Like PALB2 and BRCA2, the RAD51C gene has been evaluated as a breast and ovarian cancer susceptibility gene and as one of the causes of Fanconi anemia. Despite a possible role in German breast-ovarian cancer families and a single German family with Fanconi anemia–like features,[75,76] most studies have not found an association between RAD51C and heritable breast and ovarian cancer.[77,78] It is unclear what role this gene may play in breast and ovarian cancer susceptibility.

Genome-Wide Searches

In contrast to assessing candidate genes and/or alleles, genome wide association studies involve comparing a very large set of genetic variants spread throughout the genome. The current paradigm uses sets of 100,000 to 1 million SNPs that are chosen to capture a large portion of common variation within the genome based on the HapMap project.[79,80] By comparing allele frequencies between a large number of cases and controls, typically 1,000 or more of each, and validating promising signals in replication sets of subjects, very robust statistical signals of association have been obtained.[81-83] The strong correlation between many SNPs that are physically close to each other on the chromosome (linkage disequilibrium) allows one to “scan” the genome for susceptibility alleles even if the biologically relevant variant is not within the tested set of SNPs. While this between-SNP correlation allows one to interrogate the majority of the genome without having to assay every SNP, when a validated association is obtained, it is not usually obvious which of the many correlated variants is causal.

Genome-wide searches are showing great promise in identifying common, low-penetrance susceptibility alleles for many complex diseases,[84] including breast cancer.[85-88] The first study involved an initial scan in familial breast cancer cases followed by replication in two large sample sets of sporadic breast cancer, the final being a collection of over 20,000 cases and 20,000 controls from the BCAC, an international group of investigators.[85] Five distinct genomic regions were identified that were within or near the FGFR2, TNRC9, MAP3K1, and LSP1 genes or at the chromosome 8q region. The 8q region and others may harbor multiple independent loci associated with risk, but these regions are included only once in Table 6. Subsequent genome-wide studies have replicated these loci and identified additional ones, as summarized in Table 6.[86,87,89,89-94] SNPs identified through large studies of sporadic breast cancer appear to be associated more strongly with estrogen receptor–positive disease;[95] however, some are associated primarily or exclusively with other subtypes, including triple-negative disease.[96,97] An online catalog of SNP-trait associations from published genome-wide association studies for use in investigating genomic characteristics of trait/disease-associated SNPs (TASs) is available.

Table 6. High-probability Breast Cancer Susceptibility Loci Identified Through Genome-Wide Association Studies
Putative Gene(s) Chromosome  SNP Study Citationa Odds Ratio (OR) (95% Confidence Interval [CI])b Comments 
Intergenic /NOTCH21p11.2rs11249433[98]1.08 (1.02–1.15) [88]Stronger in ER+, low-grade [88]; also in BRCA2 [99]
ERBB2 2q34rs13393577[100]1.53 (1.37–1.70) [100]Identified in Korean subjects [100]
Intergenic2q35rs13387042[86]1.21 (1.14–1.29) [88]Stronger in bilateral and lobular [95]; also in BRCA1 and BRCA2 [101]
SLC4A7, NEK103p24rs4973768[94]1.16 (1.10–1.24) [88]Also in BRCA2 [101]
MRPS30 5p12rs10941679[93]1.11c (1.04–1.19) [88]Strongest in PR+, low-grade [102]; also in BRCA2 [101]
TERT-/CLPTM15p15rs10069690[96]1.25 (1.16–1.34) [96]Strongest in triple-negative [96]
MAP3K1 5q11.2rs889312[85]1.22 (1.14–1.30) [88]Stronger in ER+ [88]; also in BRCA2 [101]
RNF146 6q22rs2180341[89]1.24 (1.13–1.36) [103]Stronger in Ashkenazi Jews [103]
ESR1 6q25.1rs2046210[90]1.15c (1.08–1.22) [88]Also in BRCA1 [99]
TAB2 6q25.1rs9485372[104]0.90 (0.87–0.92) [104]Identified in Chinese subjects [104]
Intergenic7q32.3rs2048672[105]1.11 (1.05–1.17) [105]Identified in East Asian subjects [105]
Intergenic/MYC8q24.21rs13281615[85]1.14 (1.07–1.21) [88]Stronger in ER+ [88]
CDKN2A, CDKN2B 9p21rs1011970[88]1.09 (1.04–1.14) [88]Stronger in ER+ [88]; also in BRCA2 [106]
Intergenic9q31.2rs865686[107]0.89(0.85–0.92) [107]Also in BRCA2 [106]
ANKRD16, FBXO18 10p15.1rs2380205[88]0.94 (0.91–0.98) [88]
ZNF365 10q21.2rs10995190[88]0.86 (0.82–0.91) [88]Stronger in ER+ in general population [88]; also in BRCA2 [108]
ZMIZ1 10q22.3rs704010[88]1.07 (1.03–1.11) [88]
FGFR2 10q26.13rs2981582[85]1.43 (1.35–1.53) [88]Strongest for ER+, low-grade [95]; also in BRCA2 [101]
LSP1 11p15.5rs3817198[85]1.12 (1.05–1.19) [88]Also in BRCA2 [101]
Intergenic11q13rs614367[88]1.15 (1.10–1.20) [88]Restricted to ER+ tumors; strongest for ER+/PR+[88]
BARX211q24.3rs7107217[104]1.08 (1.05–1.11) [104]Identified in Chinese subjects [104]
PTHLH 12p11rs10771399[109]0.85 (0.83–0.88) [109]Also in BRCA1 [106]
Intergenic12q24rs1292011[109]0.92 (0.91–0.94) [109]Restricted to ER+ [109]; also in BRCA2 [106]
RAD51B 14q24.1rs999737[98]0.89 (0.83–0.95) [88]Associated with all subtypes, including triple-negative [110]; also in BRCA2 [106]
TOX3 16q12.1rs3803662[85]1.30 (1.22–1.39) [88]Stronger in ER+ [95]; also in BRCA1 and BRCA2 [101]
COX11 17q23.2rs6504950[94]0.92c (0.86–0.99) [88]
BABAM1 19p13.1rs8170[97]1.26 (1.17–1.35) [111]Restricted to triple-negative in general population [97]; also in BRCA1 [111]
NRIP1 21q21rs2823093[109]0.94 (0.92–0.96) [109]Restricted to ER+ [109]

ER- = estrogen receptor–negative; ER+ = estrogen receptor–positive; PR- = progesterone receptor–negative; PR+ = progesterone receptor–positive; SNP = single nucleotide polymorphism; triple-negative = ER-/PR-/HER2/neu-.
aInitial study that demonstrated genome-wide significance for each locus.
bAll associations observed in the general population, unless otherwise indicated; when relevant, if association was also observed in BRCA1 or BRCA2 mutation carriers, it is indicated.
cOR for best tagSNP was used [88] as a surrogate for published SNP.

Table 7. High-probability Ovarian Cancer Susceptibility Loci Identified Through Genome-Wide Association Studies
Putative Gene(s) Chromosome SNP Study Citation Odds Ratio (95% Confidence Interval) Comment 
SNP = single nucleotide polymorphism.
HOXD1 2q31.1rs2072590[112]1.16 (1.12–1.21)Stronger in serous cancers
TIPARP 3q25.31rs2665390[112]1.19 (1.11–1.27)
Intergenic/MYC, THEM758q24.21rs10088218[112]0.84 (0.80–0.89)
BNC2 9p22.2rs3814113[113]0.82 (0.79–0.86)Stronger in serous cancers; also in BRCA1 and BRCA2 [114]
SKAP1 17q21.32rs9303542[112]1.11 (1.06–1.16)
BABAM1 19p13.11rs8170[115]1.18 (1.12–1.25)Serous cancers only
ANKLE1 19p13.11rs2363956[115]1.16 (1.11–1.21)

Although the statistical evidence for an association between genetic variation at these loci and breast and ovarian cancer risk is overwhelming, the biologically relevant variants and the mechanism by which they lead to increased risk are unknown and will require further genetic and functional characterization. Additionally, these loci are associated with very modest risk (typically, odds ratio <1.5), with more risk variants likely to be identified. No interaction between the SNPs and epidemiologic risk factors for breast cancer have been identified.[116,117] At this time, because their individual and collective influences on cancer risk have not been evaluated prospectively, they are not considered clinically relevant. Furthermore, theoretical models have suggested that common moderate-risk SNPs have limited potential to improve models for individualized risk assessment.[118-120] These models used receiver operating characteristic (ROC) curve analysis to calculate the area under the curve (AUC) as a measure of discriminatory accuracy. A more recent study used ROC curve analysis to examine the utility of SNPs in a clinical dataset of greater than 5,500 breast cancer cases and nearly 6,000 controls, using a model with traditional risk factors compared to a model using both standard risk factors and ten previously identified SNPs. The addition of genetic information modestly changed the AUC from 58% to 61.8%, a result that was not felt to be clinically significant. Despite this, 32.5% of patients were in a higher quintile of breast cancer risk when genetic information was included, and 20.4% were in a lower quintile of risk. It remains unclear whether such information has clinical utility.[118,121]

More limited data are available regarding ovarian cancer risk. Three GWAS involving staged analysis of over 10,000 cases and 13,000 controls have been carried out for ovarian cancer.[112,113,115] The seven loci that reached genome-wide significance are shown in Table 7. As in other GWAS, the odds ratios are modest, generally about 1.2 or weaker, but implicate a number of genes with plausible biological ties to ovarian cancer, such as BABAM1, whose protein complexes with and may regulate BRCA1, and TIRAPR, which codes for a poly (ADP-ribose) polymerase (PARP), molecules that may be important in BRCA1/BRCA2-deficient cells.

References

  1. Easton DF: How many more breast cancer predisposition genes are there? Breast Cancer Res 1 (1): 14-7, 1999.  [PUBMED Abstract]

  2. Smith P, McGuffog L, Easton DF, et al.: A genome wide linkage search for breast cancer susceptibility genes. Genes Chromosomes Cancer 45 (7): 646-55, 2006.  [PUBMED Abstract]

  3. Pharoah PD, Antoniou A, Bobrow M, et al.: Polygenic susceptibility to breast cancer and implications for prevention. Nat Genet 31 (1): 33-6, 2002.  [PUBMED Abstract]

  4. Antoniou AC, Pharoah PP, Smith P, et al.: The BOADICEA model of genetic susceptibility to breast and ovarian cancer. Br J Cancer 91 (8): 1580-90, 2004.  [PUBMED Abstract]

  5. Chen YC, Hunter DJ: Molecular epidemiology of cancer. CA Cancer J Clin 55 (1): 45-54; quiz 57, 2005 Jan-Feb.  [PUBMED Abstract]

  6. Breast Cancer Association Consortium: Commonly studied single-nucleotide polymorphisms and breast cancer: results from the Breast Cancer Association Consortium. J Natl Cancer Inst 98 (19): 1382-96, 2006.  [PUBMED Abstract]

  7. Dunning AM, Healey CS, Pharoah PD, et al.: A systematic review of genetic polymorphisms and breast cancer risk. Cancer Epidemiol Biomarkers Prev 8 (10): 843-54, 1999.  [PUBMED Abstract]

  8. Meijers-Heijboer H, van den Ouweland A, Klijn J, et al.: Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nat Genet 31 (1): 55-9, 2002.  [PUBMED Abstract]

  9. Kuschel B, Auranen A, Gregory CS, et al.: Common polymorphisms in checkpoint kinase 2 are not associated with breast cancer risk. Cancer Epidemiol Biomarkers Prev 12 (8): 809-12, 2003.  [PUBMED Abstract]

  10. Sodha N, Bullock S, Taylor R, et al.: CHEK2 variants in susceptibility to breast cancer and evidence of retention of the wild type allele in tumours. Br J Cancer 87 (12): 1445-8, 2002.  [PUBMED Abstract]

  11. Ingvarsson S, Sigbjornsdottir BI, Huiping C, et al.: Mutation analysis of the CHK2 gene in breast carcinoma and other cancers. Breast Cancer Res 4 (3): R4, 2002.  [PUBMED Abstract]

  12. Vahteristo P, Bartkova J, Eerola H, et al.: A CHEK2 genetic variant contributing to a substantial fraction of familial breast cancer. Am J Hum Genet 71 (2): 432-8, 2002.  [PUBMED Abstract]

  13. Meijers-Heijboer H, Wijnen J, Vasen H, et al.: The CHEK2 1100delC mutation identifies families with a hereditary breast and colorectal cancer phenotype. Am J Hum Genet 72 (5): 1308-14, 2003.  [PUBMED Abstract]

  14. Schmidt MK, Tollenaar RA, de Kemp SR, et al.: Breast cancer survival and tumor characteristics in premenopausal women carrying the CHEK2*1100delC germline mutation. J Clin Oncol 25 (1): 64-9, 2007.  [PUBMED Abstract]

  15. Weischer M, Bojesen SE, Tybjaerg-Hansen A, et al.: Increased risk of breast cancer associated with CHEK2*1100delC. J Clin Oncol 25 (1): 57-63, 2007.  [PUBMED Abstract]

  16. Iniesta MD, Gorin MA, Chien LC, et al.: Absence of CHEK2*1100delC mutation in families with hereditary breast cancer in North America. Cancer Genet Cytogenet 202 (2): 136-40, 2010.  [PUBMED Abstract]

  17. Offit K, Pierce H, Kirchhoff T, et al.: Frequency of CHEK2*1100delC in New York breast cancer cases and controls. BMC Med Genet 4 (1): 1, 2003.  [PUBMED Abstract]

  18. Oldenburg RA, Kroeze-Jansema K, Kraan J, et al.: The CHEK2*1100delC variant acts as a breast cancer risk modifier in non-BRCA1/BRCA2 multiple-case families. Cancer Res 63 (23): 8153-7, 2003.  [PUBMED Abstract]

  19. Neuhausen S, Dunning A, Steele L, et al.: Role of CHEK2*1100delC in unselected series of non-BRCA1/2 male breast cancers. Int J Cancer 108 (3): 477-8, 2004.  [PUBMED Abstract]

  20. Ohayon T, Gal I, Baruch RG, et al.: CHEK2*1100delC and male breast cancer risk in Israel. Int J Cancer 108 (3): 479-80, 2004.  [PUBMED Abstract]

  21. CHEK2 Breast Cancer Case-Control Consortium.: 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 (6): 1175-82, 2004.  [PUBMED Abstract]

  22. Johnson N, Fletcher O, Naceur-Lombardelli C, et al.: Interaction between CHEK2*1100delC and other low-penetrance breast-cancer susceptibility genes: a familial study. Lancet 366 (9496): 1554-7, 2005 Oct 29-Nov 4.  [PUBMED Abstract]

  23. Fletcher O, Johnson N, Dos Santos Silva I, et al.: Family history, genetic testing, and clinical risk prediction: pooled analysis of CHEK2 1100delC in 1,828 bilateral breast cancers and 7,030 controls. Cancer Epidemiol Biomarkers Prev 18 (1): 230-4, 2009.  [PUBMED Abstract]

  24. Weischer M, Bojesen SE, Ellervik C, et al.: CHEK2*1100delC genotyping for clinical assessment of breast cancer risk: meta-analyses of 26,000 patient cases and 27,000 controls. J Clin Oncol 26 (4): 542-8, 2008.  [PUBMED Abstract]

  25. Cybulski C, Wokołorczyk D, Jakubowska A, et al.: Risk of breast cancer in women with a CHEK2 mutation with and without a family history of breast cancer. J Clin Oncol 29 (28): 3747-52, 2011.  [PUBMED Abstract]

  26. Gronwald J, Cybulski C, Piesiak W, et al.: Cancer risks in first-degree relatives of CHEK2 mutation carriers: effects of mutation type and cancer site in proband. Br J Cancer 100 (9): 1508-12, 2009.  [PUBMED Abstract]

  27. Wasielewski M, den Bakker MA, van den Ouweland A, et al.: CHEK2 1100delC and male breast cancer in the Netherlands. Breast Cancer Res Treat 116 (2): 397-400, 2009.  [PUBMED Abstract]

  28. Osorio A, Rodríguez-López R, Díez O, et al.: The breast cancer low-penetrance allele 1100delC in the CHEK2 gene is not present in Spanish familial breast cancer population. Int J Cancer 108 (1): 54-6, 2004.  [PUBMED Abstract]

  29. Syrjäkoski K, Kuukasjärvi T, Auvinen A, et al.: CHEK2 1100delC is not a risk factor for male breast cancer population. Int J Cancer 108 (3): 475-6, 2004.  [PUBMED Abstract]

  30. Tsou HC, Teng DH, Ping XL, et al.: The role of MMAC1 mutations in early-onset breast cancer: causative in association with Cowden syndrome and excluded in BRCA1-negative cases. Am J Hum Genet 61 (5): 1036-43, 1997.  [PUBMED Abstract]

  31. Olopade OI, Weber BL: Breast cancer genetics: toward molecular characterization of individuals at increased risk for breast cancer: part I. Cancer: Principles and Practice of Oncology Updates 12(10): 1-12, 1998. 

  32. Cybulski C, Górski B, Huzarski T, et al.: CHEK2-positive breast cancers in young Polish women. Clin Cancer Res 12 (16): 4832-5, 2006.  [PUBMED Abstract]

  33. Cybulski C, Huzarski T, Byrski T, et al.: Estrogen receptor status in CHEK2-positive breast cancers: implications for chemoprevention. Clin Genet 75 (1): 72-8, 2009.  [PUBMED Abstract]

  34. Offit K, Garber JE: Time to check CHEK2 in families with breast cancer? J Clin Oncol 26 (4): 519-20, 2008.  [PUBMED Abstract]

  35. Savitsky K, Bar-Shira A, Gilad S, et al.: A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268 (5218): 1749-53, 1995.  [PUBMED Abstract]

  36. Telatar M, Teraoka S, Wang Z, et al.: Ataxia-telangiectasia: identification and detection of founder-effect mutations in the ATM gene in ethnic populations. Am J Hum Genet 62 (1): 86-97, 1998.  [PUBMED Abstract]

  37. Uhrhammer N, Bay JO, Bignon YJ: Seventh International Workshop on Ataxia-Telangiectasia. Cancer Res 58 (15): 3480-5, 1998.  [PUBMED Abstract]

  38. Ahmed M, Rahman N: ATM and breast cancer susceptibility. Oncogene 25 (43): 5906-11, 2006.  [PUBMED Abstract]

  39. Khanna KK, Chenevix-Trench G: ATM and genome maintenance: defining its role in breast cancer susceptibility. J Mammary Gland Biol Neoplasia 9 (3): 247-62, 2004.  [PUBMED Abstract]

  40. Gilad S, Chessa L, Khosravi R, et al.: Genotype-phenotype relationships in ataxia-telangiectasia and variants. Am J Hum Genet 62 (3): 551-61, 1998.  [PUBMED Abstract]

  41. FitzGerald MG, Bean JM, Hegde SR, et al.: Heterozygous ATM mutations do not contribute to early onset of breast cancer. Nat Genet 15 (3): 307-10, 1997.  [PUBMED Abstract]

  42. Chen J, Birkholtz GG, Lindblom P, et al.: The role of ataxia-telangiectasia heterozygotes in familial breast cancer. Cancer Res 58 (7): 1376-9, 1998.  [PUBMED Abstract]

  43. Bay JO, Grancho M, Pernin D, et al.: No evidence for constitutional ATM mutation in breast/gastric cancer families. Int J Oncol 12 (6): 1385-90, 1998.  [PUBMED Abstract]

  44. Laake K, Vu P, Andersen TI, et al.: Screening breast cancer patients for Norwegian ATM mutations. Br J Cancer 83 (12): 1650-3, 2000.  [PUBMED Abstract]

  45. Dörk T, Bendix R, Bremer M, et al.: Spectrum of ATM gene mutations in a hospital-based series of unselected breast cancer patients. Cancer Res 61 (20): 7608-15, 2001.  [PUBMED Abstract]

  46. Teraoka SN, Malone KE, Doody DR, et al.: Increased frequency of ATM mutations in breast carcinoma patients with early onset disease and positive family history. Cancer 92 (3): 479-87, 2001.  [PUBMED Abstract]

  47. Chenevix-Trench G, Spurdle AB, Gatei M, et al.: Dominant negative ATM mutations in breast cancer families. J Natl Cancer Inst 94 (3): 205-15, 2002.  [PUBMED Abstract]

  48. Thorstenson YR, Roxas A, Kroiss R, et al.: Contributions of ATM mutations to familial breast and ovarian cancer. Cancer Res 63 (12): 3325-33, 2003.  [PUBMED Abstract]

  49. Cavaciuti E, Laugé A, Janin N, et al.: Cancer risk according to type and location of ATM mutation in ataxia-telangiectasia families. Genes Chromosomes Cancer 42 (1): 1-9, 2005.  [PUBMED Abstract]

  50. Olsen JH, Hahnemann JM, Børresen-Dale AL, et al.: Breast and other cancers in 1445 blood relatives of 75 Nordic patients with ataxia telangiectasia. Br J Cancer 93 (2): 260-5, 2005.  [PUBMED Abstract]

  51. Renwick A, Thompson D, Seal S, et al.: ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet 38 (8): 873-5, 2006.  [PUBMED Abstract]

  52. Thompson D, Duedal S, Kirner J, et al.: Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst 97 (11): 813-22, 2005.  [PUBMED Abstract]

  53. Levitus M, Waisfisz Q, Godthelp BC, et al.: The DNA helicase BRIP1 is defective in Fanconi anemia complementation group J. Nat Genet 37 (9): 934-5, 2005.  [PUBMED Abstract]

  54. Levran O, Attwooll C, Henry RT, et al.: The BRCA1-interacting helicase BRIP1 is deficient in Fanconi anemia. Nat Genet 37 (9): 931-3, 2005.  [PUBMED Abstract]

  55. Litman R, Peng M, Jin Z, et al.: BACH1 is critical for homologous recombination and appears to be the Fanconi anemia gene product FANCJ. Cancer Cell 8 (3): 255-65, 2005.  [PUBMED Abstract]

  56. Seal S, Thompson D, Renwick A, et al.: Truncating mutations in the Fanconi anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat Genet 38 (11): 1239-41, 2006.  [PUBMED Abstract]

  57. Reid S, Schindler D, Hanenberg H, et al.: Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 39 (2): 162-4, 2007.  [PUBMED Abstract]

  58. Ding YC, Steele L, Kuan CJ, et al.: Mutations in BRCA2 and PALB2 in male breast cancer cases from the United States. Breast Cancer Res Treat 126 (3): 771-8, 2011.  [PUBMED Abstract]

  59. Rahman N, Seal S, Thompson D, et al.: PALB2, which encodes a BRCA2-interacting protein, is a breast cancer susceptibility gene. Nat Genet 39 (2): 165-7, 2007.  [PUBMED Abstract]

  60. Erkko H, Dowty JG, Nikkilä J, et al.: Penetrance analysis of the PALB2 c.1592delT founder mutation. Clin Cancer Res 14 (14): 4667-71, 2008.  [PUBMED Abstract]

  61. Heikkinen T, Kärkkäinen H, Aaltonen K, et al.: The breast cancer susceptibility mutation PALB2 1592delT is associated with an aggressive tumor phenotype. Clin Cancer Res 15 (9): 3214-22, 2009.  [PUBMED Abstract]

  62. Ding YC, Steele L, Chu LH, et al.: Germline mutations in PALB2 in African-American breast cancer cases. Breast Cancer Res Treat 126 (1): 227-30, 2011.  [PUBMED Abstract]

  63. Foulkes WD, Ghadirian P, Akbari MR, et al.: Identification of a novel truncating PALB2 mutation and analysis of its contribution to early-onset breast cancer in French-Canadian women. Breast Cancer Res 9 (6): R83, 2007.  [PUBMED Abstract]

  64. Casadei S, Norquist BM, Walsh T, et al.: Contribution of inherited mutations in the BRCA2-interacting protein PALB2 to familial breast cancer. Cancer Res 71 (6): 2222-9, 2011.  [PUBMED Abstract]

  65. Southey MC, Teo ZL, Dowty JG, et al.: A PALB2 mutation associated with high risk of breast cancer. Breast Cancer Res 12 (6): R109, 2010.  [PUBMED Abstract]

  66. Hellebrand H, Sutter C, Honisch E, et al.: Germline mutations in the PALB2 gene are population specific and occur with low frequencies in familial breast cancer. Hum Mutat 32 (6): E2176-88, 2011.  [PUBMED Abstract]

  67. Bogdanova N, Sokolenko AP, Iyevleva AG, et al.: PALB2 mutations in German and Russian patients with bilateral breast cancer. Breast Cancer Res Treat 126 (2): 545-50, 2011.  [PUBMED Abstract]

  68. Wong MW, Nordfors C, Mossman D, et al.: BRIP1, PALB2, and RAD51C mutation analysis reveals their relative importance as genetic susceptibility factors for breast cancer. Breast Cancer Res Treat 127 (3): 853-9, 2011.  [PUBMED Abstract]

  69. Jones S, Hruban RH, Kamiyama M, et al.: Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 324 (5924): 217, 2009.  [PUBMED Abstract]

  70. Slater EP, Langer P, Niemczyk E, et al.: PALB2 mutations in European familial pancreatic cancer families. Clin Genet 78 (5): 490-4, 2010.  [PUBMED Abstract]

  71. Hofstatter EW, Domchek SM, Miron A, et al.: PALB2 mutations in familial breast and pancreatic cancer. Fam Cancer 10 (2): 225-31, 2011.  [PUBMED Abstract]

  72. Stadler ZK, Salo-Mullen E, Sabbaghian N, et al.: Germline PALB2 mutation analysis in breast-pancreas cancer families. J Med Genet 48 (8): 523-5, 2011.  [PUBMED Abstract]

  73. Ghiorzo P, Pensotti V, Fornarini G, et al.: Contribution of germline mutations in the BRCA and PALB2 genes to pancreatic cancer in Italy. Fam Cancer 11 (1): 41-7, 2012.  [PUBMED Abstract]

  74. Cox Angela, Dunning Alison, Garcia-Closas Montserrat, et al.: Nature genetics. Nat Genet 39 (5): 352-8, 2007. 

  75. Meindl A, Hellebrand H, Wiek C, et al.: Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene. Nat Genet 42 (5): 410-4, 2010.  [PUBMED Abstract]

  76. Vaz F, Hanenberg H, Schuster B, et al.: Mutation of the RAD51C gene in a Fanconi anemia-like disorder. Nat Genet 42 (5): 406-9, 2010.  [PUBMED Abstract]

  77. Akbari MR, Tonin P, Foulkes WD, et al.: RAD51C germline mutations in breast and ovarian cancer patients. Breast Cancer Res 12 (4): 404, 2010.  [PUBMED Abstract]

  78. Zheng Y, Zhang J, Hope K, et al.: Screening RAD51C nucleotide alterations in patients with a family history of breast and ovarian cancer. Breast Cancer Res Treat 124 (3): 857-61, 2010.  [PUBMED Abstract]

  79. The International HapMap Consortium.: The International HapMap Project. Nature 426 (6968): 789-96, 2003.  [PUBMED Abstract]

  80. Thorisson GA, Smith AV, Krishnan L, et al.: The International HapMap Project Web site. Genome Res 15 (11): 1592-3, 2005.  [PUBMED Abstract]

  81. Evans DM, Cardon LR: Genome-wide association: a promising start to a long race. Trends Genet 22 (7): 350-4, 2006.  [PUBMED Abstract]

  82. Cardon LR: Genetics. Delivering new disease genes. Science 314 (5804): 1403-5, 2006.  [PUBMED Abstract]

  83. Chanock SJ, Manolio T, Boehnke M, et al.: Replicating genotype-phenotype associations. Nature 447 (7145): 655-60, 2007.  [PUBMED Abstract]

  84. Wellcome Trust Case Control Consortium.: Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447 (7145): 661-78, 2007.  [PUBMED Abstract]

  85. Easton DF, Pooley KA, Dunning AM, et al.: Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447 (7148): 1087-93, 2007.  [PUBMED Abstract]

  86. Stacey SN, Manolescu A, Sulem P, et al.: Common variants on chromosomes 2q35 and 16q12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 39 (7): 865-9, 2007.  [PUBMED Abstract]

  87. Hunter DJ, Kraft P, Jacobs KB, et al.: A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet 39 (7): 870-4, 2007.  [PUBMED Abstract]

  88. Turnbull C, Ahmed S, Morrison J, et al.: Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet 42 (6): 504-7, 2010.  [PUBMED Abstract]

  89. Gold B, Kirchhoff T, Stefanov S, et al.: Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33. Proc Natl Acad Sci U S A 105 (11): 4340-5, 2008.  [PUBMED Abstract]

  90. Zheng W, Long J, Gao YT, et al.: Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat Genet 41 (3): 324-8, 2009.  [PUBMED Abstract]

  91. Kibriya MG, Jasmine F, Argos M, et al.: A pilot genome-wide association study of early-onset breast cancer. Breast Cancer Res Treat 114 (3): 463-77, 2009.  [PUBMED Abstract]

  92. Murabito JM, Rosenberg CL, Finger D, et al.: A genome-wide association study of breast and prostate cancer in the NHLBI's Framingham Heart Study. BMC Med Genet 8 (Suppl 1): S6, 2007.  [PUBMED Abstract]

  93. Stacey SN, Manolescu A, Sulem P, et al.: Common variants on chromosome 5p12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 40 (6): 703-6, 2008.  [PUBMED Abstract]

  94. Ahmed S, Thomas G, Ghoussaini M, et al.: Newly discovered breast cancer susceptibility loci on 3p24 and 17q23.2. Nat Genet 41 (5): 585-90, 2009.  [PUBMED Abstract]

  95. Reeves GK, Travis RC, Green J, et al.: Incidence of breast cancer and its subtypes in relation to individual and multiple low-penetrance genetic susceptibility loci. JAMA 304 (4): 426-34, 2010.  [PUBMED Abstract]

  96. Haiman CA, Chen GK, Vachon CM, et al.: A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor-negative breast cancer. Nat Genet 43 (12): 1210-4, 2011.  [PUBMED Abstract]

  97. Stevens KN, Fredericksen Z, Vachon CM, et al.: 19p13.1 is a triple-negative-specific breast cancer susceptibility locus. Cancer Res 72 (7): 1795-803, 2012.  [PUBMED Abstract]

  98. Thomas G, Jacobs KB, Kraft P, et al.: A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11.2 and 14q24.1 (RAD51L1). Nat Genet 41 (5): 579-84, 2009.  [PUBMED Abstract]

  99. Antoniou AC, Kartsonaki C, Sinilnikova OM, et al.: Common alleles at 6q25.1 and 1p11.2 are associated with breast cancer risk for BRCA1 and BRCA2 mutation carriers. Hum Mol Genet 20 (16): 3304-21, 2011.  [PUBMED Abstract]

  100. Kim HC, Lee JY, Sung H, et al.: A genome-wide association study identifies a breast cancer risk variant in ERBB4 at 2q34: results from the Seoul Breast Cancer Study. Breast Cancer Res 14 (2): R56, 2012.  [PUBMED Abstract]

  101. Antoniou AC, Beesley J, McGuffog L, et al.: Common breast cancer susceptibility alleles and the risk of breast cancer for BRCA1 and BRCA2 mutation carriers: implications for risk prediction. Cancer Res 70 (23): 9742-54, 2010.  [PUBMED Abstract]

  102. Milne RL, Goode EL, García-Closas M, et al.: Confirmation of 5p12 as a susceptibility locus for progesterone-receptor-positive, lower grade breast cancer. Cancer Epidemiol Biomarkers Prev 20 (10): 2222-31, 2011.  [PUBMED Abstract]

  103. Kirchhoff T, Chen ZQ, Gold B, et al.: The 6q22.33 locus and breast cancer susceptibility. Cancer Epidemiol Biomarkers Prev 18 (9): 2468-75, 2009.  [PUBMED Abstract]

  104. Long J, Cai Q, Sung H, et al.: Genome-wide association study in east Asians identifies novel susceptibility loci for breast cancer. PLoS Genet 8 (2): e1002532, 2012.  [PUBMED Abstract]

  105. Cai Q, Long J, Lu W, et al.: Genome-wide association study identifies breast cancer risk variant at 10q21.2: results from the Asia Breast Cancer Consortium. Hum Mol Genet 20 (24): 4991-9, 2011.  [PUBMED Abstract]

  106. Antoniou AC, Kuchenbaecker KB, Soucy P, et al.: Common variants at 12p11, 12q24, 9p21, 9q31.2 and in ZNF365 are associated with breast cancer risk for BRCA1 and/or BRCA2 mutation carriers. Breast Cancer Res 14 (1): R33, 2012.  [PUBMED Abstract]

  107. Fletcher O, Johnson N, Orr N, et al.: Novel breast cancer susceptibility locus at 9q31.2: results of a genome-wide association study. J Natl Cancer Inst 103 (5): 425-35, 2011.  [PUBMED Abstract]

  108. Couch FJ, Gaudet MM, Antoniou AC, et al.: Common variants at the 19p13.1 and ZNF365 loci are associated with ER subtypes of breast cancer and ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev 21 (4): 645-57, 2012.  [PUBMED Abstract]

  109. Ghoussaini M, Fletcher O, Michailidou K, et al.: Genome-wide association analysis identifies three new breast cancer susceptibility loci. Nat Genet 44 (3): 312-8, 2012.  [PUBMED Abstract]

  110. Figueroa JD, Garcia-Closas M, Humphreys M, et al.: Associations of common variants at 1p11.2 and 14q24.1 (RAD51L1) with breast cancer risk and heterogeneity by tumor subtype: findings from the Breast Cancer Association Consortium. Hum Mol Genet 20 (23): 4693-706, 2011.  [PUBMED Abstract]

  111. Antoniou AC, Wang X, Fredericksen ZS, et al.: A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor-negative breast cancer in the general population. Nat Genet 42 (10): 885-92, 2010.  [PUBMED Abstract]

  112. Goode EL, Chenevix-Trench G, Song H, et al.: A genome-wide association study identifies susceptibility loci for ovarian cancer at 2q31 and 8q24. Nat Genet 42 (10): 874-9, 2010.  [PUBMED Abstract]

  113. Song H, Ramus SJ, Tyrer J, et al.: A genome-wide association study identifies a new ovarian cancer susceptibility locus on 9p22.2. Nat Genet 41 (9): 996-1000, 2009.  [PUBMED Abstract]

  114. Ramus SJ, Kartsonaki C, Gayther SA, et al.: Genetic variation at 9p22.2 and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 103 (2): 105-16, 2011.  [PUBMED Abstract]

  115. Bolton KL, Tyrer J, Song H, et al.: Common variants at 19p13 are associated with susceptibility to ovarian cancer. Nat Genet 42 (10): 880-4, 2010.  [PUBMED Abstract]

  116. Campa D, Kaaks R, Le Marchand L, et al.: Interactions between genetic variants and breast cancer risk factors in the breast and prostate cancer cohort consortium. J Natl Cancer Inst 103 (16): 1252-63, 2011.  [PUBMED Abstract]

  117. Milne RL, Gaudet MM, Spurdle AB, et al.: Assessing interactions between the associations of common genetic susceptibility variants, reproductive history and body mass index with breast cancer risk in the breast cancer association consortium: a combined case-control study. Breast Cancer Res 12 (6): R110, 2010.  [PUBMED Abstract]

  118. Pharoah PD, Antoniou AC, Easton DF, et al.: Polygenes, risk prediction, and targeted prevention of breast cancer. N Engl J Med 358 (26): 2796-803, 2008.  [PUBMED Abstract]

  119. Gail MH: Discriminatory accuracy from single-nucleotide polymorphisms in models to predict breast cancer risk. J Natl Cancer Inst 100 (14): 1037-41, 2008.  [PUBMED Abstract]

  120. Gail MH: Value of adding single-nucleotide polymorphism genotypes to a breast cancer risk model. J Natl Cancer Inst 101 (13): 959-63, 2009.  [PUBMED Abstract]

  121. Wacholder S, Hartge P, Prentice R, et al.: Performance of common genetic variants in breast-cancer risk models. N Engl J Med 362 (11): 986-93, 2010.  [PUBMED Abstract]