Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Breast Dis. Author manuscript; available in PMC 2013 Dec 21.
Published in final edited form as:
PMCID: PMC3870050
NIHMSID: NIHMS496110
PMID: 21778580

BRCA1/2 MUTATIONS AND TRIPLE NEGATIVE BREAST CANCERS

Abstract

Identifying breast cancer patients at increased risk for carrying a mutation in the BRCA1 and BRCA2 genes is an important objective in clinical practice. Although age at diagnosis, family history of breast and/or ovarian cancer, and ethnicity are all essential parameters to consider when assessing risk, there are limitations as to how well such factors accurately predict BRCA1/2 status, even when quantitative risk models are applied. Integrating information about triple negative (TN) disease may help refine these estimates. Among newly diagnosed breast cancer patients, fewer than 10% have a mutation in the BRCA1 or BRCA2 genes, and up to 20% present with TN disease. However, among BRCA1 mutation carriers at least one-third have TN breast cancers. In this paper, we review key studies that have assessed breast cancer cases with a known BRCA1/2 status and triple marker data. We also discuss how integrating such information into qualitative and quantitative risk assessments of BRCA1/2 carrier probability may improve the ability to identify women who are appropriate candidates for genetic testing. Identifying women at increased risk is critical as knowledge of mutation status may impact surgical and systemic treatment in newly diagnosed patients, as well as recommendations for ovarian cancer risk management.

Keywords: BRCA1, BRCA2, genetic counseling, genetic testing, triple negative breast cancer, BRCA mutation prediction models, BRCAPRO

INTRODUCTION

Approximately 5–10% of all newly diagnosed breast cancers in Western nations are hereditary, attributable primarily to inherited mutations in the BRCA1 and BRCA2 (BRCA1/2) genes. According to a recent meta-analysis, BRCA1/2 gene mutations are associated with a 40–57% lifetime risk of female breast cancer and an 18–40% lifetime risk of ovarian cancer [10]. It is also well established that BRCA1/2 carriers with breast cancer have markedly elevated risks of contralateral breast cancer of approximately 50% at 25 years post-diagnosis [21,22].

In general, management for mutation carriers with breast cancer includes consideration of risk reducing bilateral mastectomies or alternatively, breast cancer screening with annual mammography and magnetic resonance imaging (MRI) starting at age 25 years [37]. Bilateral salpingo-oophorectomy is recommended upon completion of childbearing not only to reduce ovarian cancer risk, but when performed premenopausally, it also reduces breast cancer risk, and has been shown to reduce mortality from both of these cancers [12,37].

Newly diagnosed breast cancer patients may take advantage of rapid genetic counseling and testing to inform surgical decisions if they test positive for a BRCA1/2 mutation in that they may opt for bilateral mastectomies instead of breast conservation or unilateral mastectomy [41]. In addition, as reviewed by Rodler et al. in this volume, emerging data suggest that BRCA1/2 carriers with TN disease may respond favorably to cisplatin-targeted therapy [6,8,42]. In addition, Poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors appear to be particularly effective in BRCA1/2 mutation carriers [44], and may also play a role in the treatment of sporadic TN disease [31]. Several ongoing trials of cisplatin and PARP inhibitors in BRCA1/2 mutation carriers are currently in progress. In addition to the significant risks of developing cancer faced by probands (i.e., the first individual in a family to undergo genetic testing) with a positive test result, first-degree relatives of individuals with positive results have a 50% chance of inheriting the identified BRCA1 or BRCA2 mutation. Thus, the clinical and public health implications of identifying BRCA1/2 mutation carriers are quite significant.

Potential candidates for BRCA1/2 testing are typically identified based on a qualitative and/or quantitative analysis of personal and family history of cancer. However, given the limitations that may be encountered when interpreting a family history, increasing attention has been paid to whether breast cancer markers may provide an independent indication of women who are at increased risk of carrying a mutation. Specifically, the triple negative (TN) phenotype [i.e., defined by the absence of three markers: estrogen and progesterone receptors, and human epidermal growth factor (HER)-2 expression] is the most common histological subtype observed in BRCA1 carriers. As described below, the prevalence of TN disease in BRCA1 carriers is considerably higher than the general population prevalence of 11–20% [19].

It is important to note that many features of TN cancers overlap with those observed in basal-like cancers, but they are not identical [11]. While TN breast cancers are determined based on the assessment of the three key biomarkers mentioned above, basal-like cancers are defined based on gene expression assays and immunophenotypic profiles of cytokeratins. For the purposes of this review, we will focus primarily on TN breast cancers only, given that the pathologic markers used to determine this phenotype are routinely used in practice relative to cytokeratin studies, and therefore provide more accessible data for risk assessment.

TRIPLE NEGATIVE BREAST CANCER IN BRCA1/2 MUTATION CARRIERS

Selection of studies

A literature review was performed to identify research studies reporting on the relationship between TN breast cancer and BRCA1/2 carrier status. Studies were eligible for inclusion if the sample (or a subset of the sample) had a known BRCA1/2 mutation status and pathology sufficient to evaluate TN status. Approximately 35 publications and abstracts were reviewed on the topic. Table 1 represents the 13 key studies which were selected for inclusion, three of which were abstracts.

Table 1

Characteristics of Key Studies Assessing Relationship between Triple Negative Breast Cancer and BRCA1/2 Mutations

First author, Publication year [reference]Setting/Ascertainment criteria for BRCA1/2 genotypingSample CharacteristicsTN (n)/BRCA1 (n)TN (n)/BRCA2(n)TN (n)/BRCA1/2-(n)BRCA1 (n)/TN (n)BRCA2 (n)/TN (n)
Race/Ethnicity (%)1TN (n)/eligible sample2 (n)BRCA1(n), BRCA2 (n)BRCA1/2-(n)
Fostira 2010[18]3Athens, Greece
Unselected for age, FH; TN only
Greek/Caucasian284/284 (100%)30, n/a4254430/30 (100%)n/a4254/254 (100%)30/284 (11%)n/a4
Mavaddat 2010[34]Cambridge, UK; BCLC
Selected for BRCA1/2 status
Caucasian158/244 (65%)182, 6207143/182 (79%)515/62 (24%)5n/a6143/158 (91%)615/158 (9%)6
Saura 2010[40]3Barcelona, Spain
Selected via high risk clinic
Spanish/Caucasian58/227 (26%)20, 2718016/20 (80%)5/27 (19%)37/180 (21%)16/58 (28%)5/58 (9%)
Kwong 2009[28]Hong Kong, China
Selected via high risk clinic
Chinese59/205 (29%)712, 1771768/12 (67%)6/17 (35%)45/176 (26%)8/59 (14%)6/59 (10%)
Rakha 2009[39]Montreal, Quebec, Canada
Selected for age (≤ 65) & ethnicity (AJ)
AJ/Caucasian27/278 (10%)30, n/a4248411/26 (42%)8n/a416/248 (6%)11/27 (41%)n/a4
Yip 2009[51]Kuala Lumpur, Malaysia
Selected for BRCA1/2 risk level ≥ 10%
Chinese (62%)
Malays (26%)
Indians (13%)
26/90 (29%)10, 97110/10 (100%)3/9 (33%)13/71 (18%)10/26 (38%)3/26 (12%)
Young 2009[52]Various sites in USA
Selected for age (≤40), minimal/no FH, & ethnicity (non-AJ); TN only
Caucasian (63%)
AA (19%)
Other (15%)
Unspecified (4%)
54/54 (100%)5, 144845/5 (100%)1/1 (100%)48/48 (100%)5/54 (9%)1/54 (2%)
Atchley 2008[3]Houston, TX, USA
Selected for BRCA1/2 status9
Caucasian (72%)
Hispanic (13%)
AJ (9%)
AA (4%)
Asian (2%)
93/477 (19%)56, 3039132/56 (57%)7/30 (23%)54/391 (14%)32/93 (34%)7/93 (8%)
Li 2008[30]Various sites in China
Selected for age (< 35) or positive FH
Chinese17/78 (22%)14, n/a46447/14 (50%)n/a410/64 (16%)7/17 (41%)n/a4
Byrski 2008[7]Various sites in Poland
Selected for age (early-onset)
Polish/Caucasian22/34 (65%)34, n/a303,622/34 (65%)n/a3n/a622/22 (100%)6n/a3
Musolino 2007[35]Parma, Italy
Selected for age (<40)
Italian/Caucasian19/45 (42%)108, 311357/8 (88%)111/3 (33%)1112/35 (34%)7/19 (37%)1/19 (5%)
Haffty 2006[23]New Haven, CT, USA
Selected for BRCA1/2 status
Caucasian (83%)
AA (13%)
Other (4%)
34/99 (34%)10, 7828/10 (80%)1/7 (14%)25/82 (30%)8/34 (24%)1/34 (3%)
Kandel 2006[26]3Boston, MA, USA; SPORE
Selected for BRCA1/2 status; TN only
Unspecified23/23 (100%)9, 2129/9 (100%)2/2 (100%)12/12 (100%)9/23 (39%)2/23 (9%)

Abbreviations: ER−: estrogen receptor negative; PR−: progesterone receptor negative; HER-2 neg: human epidermal growth factor receptor 2 negative; TN: triple negative= ER−, PR−, and HER-2neg; BRCA1: BRCA1 mutation carrier; BRCA2: BRCA2 mutation carrier; BRCA1/2−: negative for mutations in these genes; FH: family history of breast and/or ovarian cancer; n/a: not applicable; BCLC: Breast Cancer Linkage Consortium; AJ: Ashkenazi Jewish; AA: African American; SPORE: Specialized Programs of Research Excellence

1Percentages may not add to 100% due to rounding
2Eligible sample = BRCA status known + pathology available to determine TN status
3Abstract data only
4Some participants had incomplete BRCA1/2 testing (e.g. only BRCA1, only founder mutations, or BRCA2 data not reported in original study)
5TN proportions only calculated among cases known to be ER −
6Sample consisted of known BRCA1/2 carriers only
7Eligible sample refers to number of tumors; includes 9 bilateral cases (4 BRCA1, 5 BRCA2)
8This figure may be an underestimate. If the 19 BRCA1 tumors that were ER− and HER-2 negative were also PR− (data not available), this proportion would be 19/29 or 66% (see table 3 in ref [39] )
9Original study excluded variants and double heterozygote from analysis
10TN = ER−, PR−, HER-2 negative, and p53+
11Double heterozygote included with BRCA1 and BRCA2 carrier

Proportion of TN Breast Cancer by BRCA1/2 Status

Excluding studies in which all cases had TN breast cancer, the proportion of TN breast cancer among BRCA1 carriers ranged from 42–100%. The lower end of this range derived from the paper by Rakha et al. [39] is likely to be an underestimate given that some of the tumors acquired in that study were assessed for only two markers (ER and HER-2) and were negative; however, it is likely that many of these tumors were PR− as well, which would mean that up to 66% of the tumors in BRCA1 carriers were TN. On the other hand, the proportion of TN breast cancer among known BRCA2 carriers and BRCA1/2 non-carriers both tended to be lower and more similar to each other (14–35% and 6–34%, respectively). Excluding studies that selected only TN breast cancers, the overall proportion of those that were TN was highest among BRCA1 carriers, at 71%. The overall proportions of breast cancer cases that were TN among BRCA2 carriers and BRCA1/2 non-carriers were 25% and 17%, respectively.

Prevalence of BRCA1/2 Carriers among TN Breast Cancer Cases

The prevalence of mutation carriers among patients with TN breast cancer is more variable than the TN prevalence among known carriers, as would be expected. In studies that calculated the prevalence of both BRCA1 mutations and BRCA2 mutations among TN breast cancer cases, the prevalence of BRCA1 mutations was always higher. The proportion of BRCA1 mutations among TN cases ranged from 9–100% (mean 35%), while the proportion of BRCA2 mutations among TN cases ranged from 2–12% (mean 8%). The range among BRCA1 carriers is wider than that for the BRCA2 carriers; however, it is again important to consider outliers in this trend, which may be attributable to selection criteria of specific studies. For example, in the study by Young et al. (2009) [52], cases were selected based on age at breast cancer diagnosis only (i.e., age 40 or younger) with minimal family history. Thus, the number of BRCA1/2 carriers is very low.

Considerations in Ascertainment

When assessing the relationship between BRCA1/2 carrier status and TN disease, it is important to consider limitations in ascertainment criteria of the various studies. For example, two studies selected participants who were known BRCA1/2 carriers [7,34], 3 studies selected patients from high risk clinics or based on a high prior probability of carrying a mutation [28,40,51], and 3 studies selected only participants with known TN breast cancer cases [18,26,52]. In 6 studies, BRCA1/2 testing was performed prospectively [18,28,30,35,51,52]. Studies that included participants who underwent BRCA1/2 testing because of established risk factors were more likely to have higher proportions of carriers, as one would expect. Fostira et al. [18] performed prospective testing regardless of risk level (i.e., unselected for family history, etc.), whereas the remaining 5 studies with prospective testing selected individuals for testing based on established risk factors (e.g., age at diagnosis and/or family history) [28,30,35,51,52]. Likewise, the 7 studies that assessed retrospective BRCA1/2 test results had variable selection criteria. Consequently, this may account for some variability in the proportion of BRCA1 carriers among TN cases, as some individuals were more likely to test positive based on the presence of specific risk factors other than the tumor pathology. Moreover, in retrospective studies, in which patients were tested many years after their diagnosis, survivorship bias may affect prevalence rates. For example, if BRCA1 carriers with TN disease had a higher rate of mortality relative to women with non-TN disease, then prevalence rates of carriers would be underestimated in the TN group and over-estimated in the non-TN group.

The majority of studies identified were in Caucasians; however, several studies were conducted in Chinese, various sub-populations of Caucasians (e.g., Ashkenazi Jewish), and in samples where race/ethnicity was not reported. The limited information regarding the relationship between TN breast cancer and BRCA1/2 status in various ethnicities/races makes it difficult to assess for specific trends using this parameter.

Other limitations noted in some of the studies selected in this review include small sample sizes, variability in the extent of genetic testing performed, variability in how pathology was assessed, and absence of data on BRCA1/2 non-carriers.

IDENTIFICATION OF BRCA1/2 MUTATION CARRIERS

One of the challenges faced by clinicians is the identification of individuals for whom genetic testing is appropriate. Several key factors are used to aid in this process, including the age at diagnosis for the proband (e.g., breast cancer diagnosed < 50 years); ethnicity (e.g., presence of Ashkenazi Jewish ancestry); and family history, with the presence of epithelial ovarian cancer being a strong predictor of identifying a BRCA1/2 mutation [17]. Increasingly, integration of breast cancer pathology, particularly TN status, is being used to refine estimates of BRCA1 carrier probability.

Most genetic testing guidelines consist of criteria that include ethnicity as well as an individual’s personal and family history of breast and genetically associated cancers, such as ovarian, prostate, and pancreatic [1,37,45]. However, as age of breast cancer diagnosis alone may be an indicator of hereditary risk, it is appropriate to offer BRCA1/2 testing to young affected breast cancer patients even if they have no or minimal family history [37]. In many instances, though, these women will not test positive for BRCA1/2 mutations [5,20,53]. In evaluating candidates for genetic testing, clinicians need to consider whether family history is limited or adequate enough to properly assess hereditary cancer risk [50].

Quantitative models are often useful adjuncts to qualitative pedigree interpretations to provide mathematical estimates of BRCA1/2 carrier probability and associated a priori risks of breast and ovarian cancer, although such models have varying degrees of discriminatory accuracy [2,33,38]. Commonly used models in current clinical practice to assess probands with breast cancer include Myriad data [36], Penn II [47], BRCAPRO [4], BOADICEA [46], and Manchester [13,15]. These models vary in how they consider personal and family history of breast, ovarian, and other cancers; age at diagnosis of affected individuals; presence of and age of unaffected relatives; oophorectomy status; bilaterality of breast cancer; race/ethnicity; gene mutation penetrance; and contribution of genes other than BRCA1/2 to hereditary breast cancer.

As mentioned, it is increasingly recognized that incorporating breast cancer pathology may improve BRCA1/2 carrier risk prediction, particularly for breast cancer patients with less striking family histories. Specifically, the histopathology and immunohistochemistry of BRCA1-associated breast cancers may deviate in notable ways from patterns observed in sporadic (i.e., non-hereditary) cases [24]. Comparable to women with sporadic breast cancer, the most frequent histopathology in BRCA1 mutation carriers is invasive ductal adenocarcinoma; however, medullary carcinoma and atypical medullary carcinoma are over-represented in BRCA1 carriers. As previously discussed, the tumors in BRCA1 carriers are more frequently high grade and are negative for estrogen and progesterone receptors, and HER-2. Additionally, a high proportion of BRCA1 tumors express basal markers such as the high molecular weight cytokeratins (CK), including CK5/6 and CK14 [24], although these markers are not currently routinely assessed in clinical practice. Conversely, studies have not demonstrated consistent findings regarding whether BRCA2-associated breast cancers exhibit a different histological profile relative to sporadic cases; however, they are more frequently moderately to poorly differentiated owing to less tubule formation, and have a higher proportion of continuous pushing margins [24]. Molecular profiles with regard to hormone receptor status, HER-2 protein expression, and basal markers in BRCA2-associated breast cancers appear to be similar to profiles observed in sporadic cases [24].

Thus, given the unique molecular phenotype noted in BRCA1 carriers with breast cancer, in particular, the high prevalence of TN tumors, such information could be useful in improving a priori carrier prediction. Several studies have demonstrated that the use of tumor morphology, namely ER status and histological grade, can increase the sensitivity of BRCA1 predictions even when family history is not considered [9,16,29,32]. Not surprisingly, more recent studies have shown that the inclusion of TN and basal markers is likely to be highly predictive of BRCA1 status, particularly when basal markers are assessed in TN tumors [49].

Further, quantitative models have begun to integrate breast tumor markers in assessing BRCA carrier probabilities. Mavaddat et al. [34] used several tumor characteristics (e.g., receptor and triple negative status, basal markers) to improve the accuracy of BOADICEA. This modification is expected to become available on the BOADICEA web interface [34]. The Manchester scoring system was recently modified to include several pathological features, including grade 3 triple negativity, which significantly improved the model’s ability to predict BRCA1 status within a validation study [14]. The other model, which is the most widely used in the United States, BRCAPRO, considers positive or negative status for the following markers: ER, PR, and cytokeratins (CK5/6 and CK14) [43]. In a recent study, BRCAPRO outperformed five other models in selecting individuals for BRCA1/2 mutation testing in clinical practice [25]. However, using tumor grade and hormone receptor status for an affected proband significantly improved the ability to discriminate BRCA1 positive families, but not BRCA2 positive families [25].

Finally, a recent Markov Monte Carlo simulation demonstrated that performing BRCA1/2 testing in women with TN breast cancers diagnosed younger than age 50 is cost-effective, and if systematically applied in the U.S., could reduce the number of new breast cancers by 23% and reduce the number ovarian cancers by 41% [27]. The authors estimate that only 7–10% of unselected women with breast cancer will test positive for a BRCA1 or BRCA2 mutation, whereas up to 25% of women with TN breast cancer unselected for family history will test positive [27].

As an example of how consideration of TN status affects carrier probability, Figure 1 shows a hypothetical pedigree of a newly diagnosed breast cancer patient who presents for genetic counseling. She was diagnosed with breast cancer at age 40 and her mother was diagnosed with breast cancer at age 45. The pedigree was entered into CancerGene version 5.1 [48]. This computing platform includes the BRCAPRO model, which considers tumor pathology in addition to family history to calculate BRCA1/2 carrier probabilities. As mentioned, the breast cancer tumor parameters that can be entered are ER, PR, CK5/6 and CK14 status (not tested/unknown, positive, or negative). Of note, the BRCAPRO model does not consider HER-2 status because it was not shown to be predictive of tumor marker status after accounting for ER [43]. When all markers are unknown, the proband’s probability of testing positive for a BRCA1 or BRCA2 mutation is only 3.6%. However, when her negative ER and PR status is entered, the carrier probability rises to 10.3% (i.e., 8.5% chance of a BRCA1 mutation specifically), and when positive CK status for both markers is also included, her probability is a remarkable 50.6% (with most of that risk, 49.6%, accounting for a BRCA1 mutation).

An external file that holds a picture, illustration, etc.
Object name is nihms496110f1.jpg

Pedigree of newly diagnosed breast cancer patient

In summary, the integration of extensive tumor pathology including TN and basal markers appears to improve the ability to predict BRCA1 carrier probability compared to standard risk factor analysis. In some cases, the resulting change in risk assessment with molecular markers could influence recommendations for genetic testing or the patient’s decision to pursue genetic testing. It is also possible that consideration of molecular markers will become formally integrated into genetic testing criteria, including those used by insurance companies. However, it is important to note that at the present time, while markers for TN status are routinely assessed, CK markers are not.

SUMMARY

The ability to identify women at increased risk for carrying a BRCA1 or BRCA2 mutation remains an important priority for clinicians and researchers. In addition to traditional parameters used in risk assessment, such as age at cancer diagnosis, family history, and ethnicity, the addition of pathological markers often improves prediction of carrier probability. Specifically, the recognition that BRCA1 carriers are more likely to develop TN breast cancers and express basal markers are important considerations in risk assessment, particularly for patients with less striking family histories. Quantitative models of BRCA1/2 probability are increasingly integrating breast tumor pathology and are expected to undergo continued validation in the future. Once BRCA1/2 mutation carriers are identified, management focuses on reducing the risk of breast and ovarian cancer, and extending genetic counseling and testing to interested at-risk relatives. Although knowledge of their BRCA1/2 status may already influence surgical decision making in newly diagnosed breast cancer patients, such information may eventually play a more central role in systemic treatment decisions as well.

Acknowledgments

The authors thank Susan Marx for assistance in preparing the manuscript.

Footnotes

Disclaimer: None of the authors has a financial conflict of interest. This research was supported in part by the Jess and Mildred Fisher Center for Familial Cancer Research, Georgetown University, Lombardi Comprehensive Cancer Center.

Contributor Information

Beth N. Peshkin, Fisher Center for Familial Cancer Research, Georgetown University, Lombardi Comprehensive Cancer Center, 3300 Whitehaven Street, NW, Suite 4100, Washington, DC 20007-2401, Phone: 202.687.2716, Fax: 202.687.0305.

Michelle L. Alabek, Norton Cancer Institute, 3991 Dutchmans Lane, Suburban Plaza II, Suite 405, Louisville, KY 40207, Phone: 502.899.6818, Fax: 502.899.6763.

Claudine Isaacs, Fisher Center for Familial Cancer Research, Georgetown University, Lombardi Comprehensive Cancer Center, 3800 Reservoir Road, NW, Washington, DC 20007, Phone: 202.444.3677, Fax: 202. 444.9429.

References

1. American College of Obstetricians and Gynecologists, ACOG Committee on Practice Bulletins, ACOG Committee on Genetics, Society of Gynecologic Oncologists. ACOG Practice Bulletin No. 103: Hereditary breast and ovarian cancer syndrome. Obstet Gynecol. 2009;113:957–966. [PubMed] [Google Scholar]
2. Antoniou AC, Hardy R, Walker L, et al. Predicting the likelihood of carrying a BRCA1 or BRCA2 mutation: validation of BOADICEA, BRCAPRO, IBIS, Myriad and the Manchester scoring system using data from UK genetics clinics. J Med Genet. 2008;45:425–431. [PubMed] [Google Scholar]
3. Atchley DP, Albarracin CT, Lopez A, et al. Clinical and pathologic characteristics of patients with BRCA-positive and BRCA-negative breast cancer. J Clin Oncol. 2008;26:4282–4288. [PMC free article] [PubMed] [Google Scholar]
4. BayesMendel Lab at Johns Hopkins Medicine. BRCAPRO. Available at http://astor.som.jhmi.edu/BayesMendel/brcapro.html.
5. Bonadona V, Sinilnikova OM, Chopin S, et al. Contribution of BRCA1 and BRCA2 germ-line mutations to the incidence of breast cancer in young women: results from a prospective population-based study in France. Genes Chromosomes Cancer. 2005;43:404–413. [PubMed] [Google Scholar]
6. Byrski T, Gronwald J, Huzarski T, et al. Pathologic complete response rates in young women with BRCA1-positive breast cancers after neoadjuvant chemotherapy. J Clin Oncol. 2010;28:375–379. [PubMed] [Google Scholar]
7. Byrski T, Gronwald J, Huzarski T, et al. Response to neo-adjuvant chemotherapy in women with BRCA1-positive breast cancers. Breast Cancer Res Treat. 2008;108:289–296. [PubMed] [Google Scholar]
8. Byrski T, Huzarski T, Dent R, et al. Response to neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat. 2009;115:359–363. [PubMed] [Google Scholar]
9. Chang J, Hilsenbeck SG, Sng JH, Wong J, Ragu GC. Pathological features and BRCA1 mutation screening in premenopausal breast cancer patients. Clin Cancer Res. 2001;7:1739–1742. [PubMed] [Google Scholar]
10. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol. 2007;25:1329–1333. [PMC free article] [PubMed] [Google Scholar]
11. Dawson SJ, Provenzano E, Caldas C. Triple negative breast cancers: clinical and prognostic implications. Eur J Cancer. 2009;45(Suppl 1):27–40. [PubMed] [Google Scholar]
12. Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA. 2010;304:967–975. [PMC free article] [PubMed] [Google Scholar]
13. Evans DG, Eccles DM, Rahman N, et al. A new scoring system for the chances of identifying a BRCA1/2 mutation outperforms existing models including BRCAPRO. J Med Genet. 2004;41:474–480. [PMC free article] [PubMed] [Google Scholar]
14. Evans DG, Lalloo F, Cramer A, et al. Addition of pathology and biomarker information significantly improves the performance of the Manchester scoring system for BRCA1 and BRCA2 testing. J Med Genet. 2009;46:811–817. [PubMed] [Google Scholar]
15. Evans DG, Lalloo F, Wallace A, Rahman N. Update on the Manchester Scoring System for BRCA1 and BRCA2 testing. J Med Genet. 2005;42:e39. [PMC free article] [PubMed] [Google Scholar]
16. Farshid G, Balleine RL, Cummings M, Waring P. Kathleen Cuningham Consortium for Research into Familial Breast Cancer (kConFab), Morphology of breast cancer as a means of triage of patients for BRCA1 genetic testing. Am J Surg Pathol. 2006;30:1357–1366. [PubMed] [Google Scholar]
17. Ford D, Easton DF, Stratton M, et al. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. Am J Hum Genet. 1998;62:676–689. [PMC free article] [PubMed] [Google Scholar]
18. Fostira F, Tsitlaidou M, Gogas H, et al. Prevalence of BRCA1 mutations among 284 women with triple-negative breast cancer. 2010 ASCO Annual Meeting (abstract 1511) J Clin Oncol. 28(15 Suppl) Published online http://abstract.asco.org/AbstView_74_49840.html. [Google Scholar]
19. Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, Harbeck N. Triple-negative breast cancer--current status and future directions. Ann Oncol. 2009;20:1913–1927. [PubMed] [Google Scholar]
20. Golshan M, Miron A, Nixon AJ, et al. The prevalence of germline BRCA1 and BRCA2 mutations in young women with breast cancer undergoing breast-conservation therapy. Am J Surg. 2006;192:58–62. [PubMed] [Google Scholar]
21. Graeser MK, Engel C, Rhiem K, et al. Contralateral breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Clin Oncol. 2009;27:5887–5892. [PubMed] [Google Scholar]
22. Gronwald J, Tung N, Foulkes WD, et al. Tamoxifen and contralateral breast cancer in BRCA1 and BRCA2 carriers: an update. Int J Cancer. 2006;118:2281–2284. [PubMed] [Google Scholar]
23. Haffty BG, Yang Q, Reiss M, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24:5652–5657. [PubMed] [Google Scholar]
24. Honrado E, Benitez J, Palacios J. Histopathology of BRCA1- and BRCA2-associated breast cancer. Crit Rev Oncol Hematol. 2006;59:27–39. [PubMed] [Google Scholar]
25. James PA, Doherty R, Harris M, et al. Optimal selection of individuals for BRCA mutation testing: a comparison of available methods. J Clin Oncol. 2006;24:707–715. [PubMed] [Google Scholar]
26. Kandel MJ, Stadler Z, Masciari S, et al. Prevalence of BRCA1 mutations in triple negative breast cancer (BC). 2006 ASCO Annual Meeting Proceedings Part I [abstract 508] J Clin Oncol. 2006;24(18S - June 20 Suppl) Published online http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=40&abstractID=34683. [Google Scholar]
27. Kwon JS, Gutierrez-Barrera AM, Young D, et al. Expanding the criteria for BRCA mutation testing in breast cancer survivors. J Clin Oncol. 2010;28:4214–4220. [PubMed] [Google Scholar]
28. Kwong A, Wong LP, Wong HN, et al. Clinical and pathological characteristics of Chinese patients with BRCA related breast cancer. Hugo J. 2009;3:63–76. [PMC free article] [PubMed] [Google Scholar]
29. Lakhani SR, Van De Vijver MJ, Jacquemier J, et al. The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol. 2002;20:2310–2318. [PubMed] [Google Scholar]
30. Li WF, Hu Z, Rao NY, et al. The prevalence of BRCA1 and BRCA2 germline mutations in high-risk breast cancer patients of Chinese Han nationality: two recurrent mutations were identified. Breast Cancer Res Treat. 2008;110:99–109. [PubMed] [Google Scholar]
31. Liang H, Tan AR. Iniparib, a PARP1 inhibitor for the potential treatment of cancer, including triple-negative breast cancer. IDrugs. 2010;13:646–656. [PubMed] [Google Scholar]
32. Lidereau R, Eisinger F, Champeme MH, et al. Major improvement in the efficacy of BRCA1 mutation screening using morphoclinical features of breast cancer. Cancer Res. 2000;60:1206–1210. [PubMed] [Google Scholar]
33. Lindor NM, Johnson KJ, Harvey H, et al. Predicting BRCA1 and BRCA2 gene mutation carriers: comparison of PENN II model to previous study. Fam Cancer. 2010 [PMC free article] [PubMed] [Google Scholar]
34. Mavaddat N, Rebbeck TR, Lakhani SR, Easton DF, Antoniou AC. Incorporating tumour pathology information into breast cancer risk prediction algorithms. Breast Cancer Res. 2010;12:R28. [PMC free article] [PubMed] [Google Scholar]
35. Musolino A, Bella MA, Bortesi B, et al. BRCA mutations, molecular markers, and clinical variables in early-onset breast cancer: a population-based study. Breast. 2007;16:280–292. [Google Scholar]
36. Myriad Genetic Laboratories, Inc. Mutation prevalence tables. Available at http://www.myriad.com/lib/brac/brca-prevalence-tables.pdf.
37. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast and ovarian, V.I.2010. Available at http://www.nccn.org/professionals/physician_gls/PDF/genetics_screening.pdf.
38. Parmigiani G, Chen S, Iversen ES, Jr, et al. Validity of models for predicting BRCA1 and BRCA2 mutations. Ann Intern Med. 2007;147:441–450. [PMC free article] [PubMed] [Google Scholar]
39. Rakha EA, Elsheikh SE, Aleskandarany MA, et al. Triple-negative breast cancer: distinguishing between basal and nonbasal subtypes. Clin Cancer Res. 2009;15:2302–2310. [PubMed] [Google Scholar]
40. Saura C, Sanchez-Olle G, Bosch N, et al. High prevalence of BRCA1/2 germline mutations in female breast cancer patients with triple-negative phenotype (TNBC) and family history. 2010 ASCO Annual Meeting [abstract 1534] J Clin Oncol. 2010;28(15 Suppl) Published online http://abstract.asco.org/AbstView_74_52870.html. [Google Scholar]
41. Schwartz MD, Lerman C, Brogan B, et al. Impact of BRCA1/BRCA2 counseling and testing on newly diagnosed breast cancer patients. J Clin Oncol. 2004;22:1823–1829. [PubMed] [Google Scholar]
42. Silver DP, Richardson AL, Eklund AC, et al. Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol. 2010;28:1145–1153. [PMC free article] [PubMed] [Google Scholar]
43. Tai YC, Chen S, Parmigiani G, Klein AP. Incorporating tumor immunohistochemical markers in BRCA1 and BRCA2 carrier prediction. Breast Cancer Res. 2008;10:401. [PMC free article] [PubMed] [Google Scholar]
44. Tutt A, Robson M, Garber JE, et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376:235–244. [PubMed] [Google Scholar]
45. U.S. Preventive Services Task Force. Genetic risk assessment and BRCA mutation testing for breast and ovarian cancer susceptibility: recommendation statement. Ann Intern Med. 2005;143:355–361. [PubMed] [Google Scholar]
46. University of Cambridge and Genetic Epidemiology Unit. BOADICEA. Available at http://www.srl.cam.ac.uk/genepi/boadicea/boadicea_home.html.
47. University of Pennsylvania Abramson Cancer Center. The Penn II BRCA1 and BRCA2 mutation risk evaluation model official web site. Available at http://www.afcri.upenn.edu/itacc/penn2/
48. University of Texas Southwestern Medical Center at Dallas. CancerGene [computer program] Available at http://www.utsouthwestern.edu/utsw/cda/dept47829/files/65844.html.
49. Vargas AC, Silva LD, Lakhani SR. The contribution of breast cancer pathology to statistical models to predict mutation risk in BRCA carriers. Fam Cancer. 2010 [PubMed] [Google Scholar]
50. Weitzel JN, Lagos VI, Cullinane CA, et al. Limited family structure and BRCA gene mutation status in single cases of breast cancer. JAMA. 2007;297:2587–2595. [PubMed] [Google Scholar]
51. Yip CH, Taib NA, Choo WY, Rampal S, Thong MK, Teo SH. Clinical and pathologic differences between BRCA1-, BRCA2-, and non-BRCA-associated breast cancers in a multiracial developing country. World J Surg. 2009;33:2077–2081. [PubMed] [Google Scholar]
52. Young SR, Pilarski RT, Donenberg T, et al. The prevalence of BRCA1 mutations among young women with triple-negative breast cancer. BMC Cancer. 2009;9:86. [PMC free article] [PubMed] [Google Scholar]
53. Zakhartseva LM, Gorovenko NG, Podolskaya SV, et al. Breast cancer immunohistochemical features in young women with BRCA 1/2 mutations. Exp Oncol. 2009;31:174–178. [PubMed] [Google Scholar]
-