ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Cancer risks and management of BRCA1/2 carriers without cancer

Cancer risks and management of BRCA1/2 carriers without cancer
Literature review current through: Jan 2024.
This topic last updated: Jan 29, 2024.

INTRODUCTION — Mutations in either of the breast cancer type 1 or 2 susceptibility genes (BRCA1 and BRCA2; referred in this topic as BRCA1/2) account for the majority of hereditary breast and ovarian cancers with an identified pathogenic variant in a cancer susceptibility gene. Overall, pathogenic variants in these genes are implicated in about 15 percent of women with familial breast cancer and a similar proportion of all women with incident ovarian cancers [1,2].

Hereditary breast and ovarian cancer attributable to pathogenic variants in BRCA1/2 is characterized by an autosomal-dominant pattern of inheritance, markedly increased susceptibility to breast and ovarian cancer, with an especially early onset of breast cancer, and an increased incidence of tumors of other organs, such as the fallopian tubes, prostate, male breast, and pancreas. This topic discusses the management of patients with pathogenic variants in BRCA1/2.

The selection of patients who should be offered genetic risk evaluation is discussed elsewhere. Management for individuals with well-defined, high-risk syndromes that are not due to BRCA1/2 (eg, Li-Fraumeni syndrome and phosphatase and tensin homolog [PTEN] hamartoma tumor syndrome) is also discussed separately.

(See "Genetic testing and management of individuals at risk of hereditary breast and ovarian cancer syndromes", section on 'Criteria for genetic risk evaluation'.)

(See "Overview of hereditary breast and ovarian cancer syndromes".)

Finally, management of patients with germline BRCA1/2 mutations and a diagnosis of cancer are discussed as follows:

Breast cancer

(See "Contralateral prophylactic mastectomy", section on 'BRCA carriers'.)

(See "Breast cancer in men".)

(See "ER/PR negative, HER2-negative (triple-negative) breast cancer", section on 'Germline BRCA mutation'.)

(See "Overview of the approach to metastatic breast cancer", section on 'Special considerations'.)

Ovarian cancer

(See "Approach to survivors of epithelial ovarian, fallopian tube, or peritoneal carcinoma", section on 'Breast cancer risk management in BRCA mutation carriers with EOC'.)

(See "Medical treatment for relapsed epithelial ovarian, fallopian tube, or peritoneal cancer: Platinum-sensitive disease", section on 'PARP inhibition in BRCA carriers'.)

(See "Medical treatment for relapsed epithelial ovarian, fallopian tube, or peritoneal cancer: Platinum-resistant disease", section on 'Patients with a BRCA mutation'.)

CLINICAL CHARACTERISTICS ASSOCIATED WITH BRCA1/2 PATHOGENIC VARIANTS — Most hereditary breast and ovarian cancers are due to highly penetrant germline BRCA1/2 pathogenic variants, which are inherited in an autosomal-dominant fashion. The prevalence of germline BRCA1/2 pathogenic variants is increased in certain ethnic or racial groups such as women of Ashkenazi Jewish ethnicity. (See 'Prevalence of BRCA1/2 pathogenic variants' below.)

In patients with BRCA1/2 pathogenic variants, there are frequently several generations of women affected with breast cancer (often premenopausal), as well as, in some families, ovarian cancer. In addition, other BRCA1/2-associated malignancies such as prostate, male breast, and pancreatic cancer may be observed. Specific criteria for genetic evaluation in high-risk individuals are discussed separately. (See "Genetic testing and management of individuals at risk of hereditary breast and ovarian cancer syndromes", section on 'Criteria for genetic risk evaluation'.)

PREVALENCE OF BRCA1/2 PATHOGENIC VARIANTS — The prevalence of BRCA1/2 pathogenic variants varies based on a number of factors, including ethnicity and, for those with cancer, age at diagnosis. Several founder mutations (ie, particular BRCA1/2 mutations occurring among defined ethnic groups or individuals from a specific geographic area) have been observed. Aside from Ashkenazi Jews, founder mutations have also been reported worldwide in populations from the Netherlands, Sweden, Hungary, Iceland, Italy, France, South Africa, Pakistan, Asia, and among French Canadians, Latinx, and African Americans [3-8].

Ashkenazi Jewish – In an unselected non-Jewish population in the United States, the chance of having any pathogenic variant in BRCA1/2 is about 1 in 400 [9]. By comparison, in Ashkenazi Jews unselected for personal/family cancer history (from Central or Eastern Europe), roughly 1 in 40 individuals (2.5 percent) has one of three founder mutations: 185delAG (also known as 187delAG or c.68_69delAG in BRCA1), 5382insC (also known as 5385insc or c.5266dupC in BRCA1), or 6174delT (c.5946delT) in BRCA2 [10-13]. These three pathogenic variants account for about 90 percent of BRCA1/2 mutations identified in this ethnic group [14-17].

Icelandic – A founder mutation in BRCA2, 999del5, is present in approximately 8 percent of female breast cancer cases, 40 percent of male breast cancer cases, and 6 percent of ovarian cancer cases in Iceland [18,19]. It is present in 0.6 percent of the general Icelandic population [18]. Another founder mutation in BRCA1, G5193A, has also been identified, but the prevalence of this mutation is very low [19,20].

French Canadian – At least three BRCA1 and three BRCA2 founder mutations account for about 75 to 85 percent of mutations present in French Canadians [21]. In an analysis of 1093 French Canadian women with noninvasive or invasive breast cancer, the prevalence of a founder mutation was 5.3 percent for women under age 50 years and 4.6 percent for women over age 50 with at least two affected first- or second-degree relatives with breast cancer [22]. The penetrance of BRCA2 mutations is high relative to most other populations [21].

Latinx – Nine pathogenic variants account for 53 percent of the BRCA1/2 mutations identified in the Hispanic population in the United States, with the Ashkenazi Jewish founder mutation 185delAG (187delAG) being the most common [23].

African American – Historically, the frequency of pathogenic BRCA1/2 variants in patients of African American ancestry was reported to be low [24-28]. However, among 947 individuals of African descent referred for clinical molecular testing for suspicion of hereditary breast and ovarian cancer but who did not meet stringent high-risk criteria, 11.5 percent tested positive for a BRCA1/2 mutation; among 1767 African individuals meeting stringent high-risk criteria, 29.4 percent tested positive for a BRCA1/2 mutation, including 43 (2.4 percent) who had a large rearrangement [8]. Interestingly, several specific BRCA1 large rearrangements were detected in this heterogeneous population of African ancestry, including a duplication of exons 18 to 19 in BRCA1.

CANCER RISKS IN BRCA1/2 CARRIERS — Patients with a pathogenic BRCA1/2 variant are at an increased risk of breast, ovarian, and other cancers. These risks are not isolated to women, but also affect male carriers. A 2022 international study of 3184 BRCA1 carriers and 2157 BRCA2 carriers reported on age-specific absolute risks for 22 cancer types.

The primary cancer risks are discussed below.

Breast and ovarian cancer — In clinical practice, it is difficult to provide highly individualized predictions about cancer risks. Therefore, we often provide patients with a range of risks and then tailor risk estimates to the implicated gene, their current age, breast cancer history, use of hormonal therapy, and oophorectomy status. Common genetic variations, called single-nucleotide polymorphisms (SNPs), may also contribute significantly to risk, particularly in BRCA2 carriers; however, these are generally not assessed in routine clinical practice.

The range of risks that we quote is based on three meta-analyses [5,29,30] and two prospective studies [5,31].

We quote the range of lifetime breast cancer risk in BRCA1 carriers to be between 57 and 72 percent, and between 45 and 69 percent in BRCA2 carriers. We quote the lifetime risk for ovarian cancer to be between 39 and 59 percent for BRCA1 carriers and 11 to 20 percent for BRCA2 carriers. The risk of ovarian cancer under the age of 40 years, however, is low [5,29,31,32].

As an example of lifetime risk of breast and ovarian cancers, one contemporary prospective cohort study included 9856 patients with a pathogenic variant in BRCA1/2 (approximately 60 and 40 percent for BRCA1 and BRCA2, respectively) and reported the cumulative cancer risks to age 80 years as follows [31]:

BRCA1 mutation carriers

Breast – 72 percent (95% CI 65 to 79 percent)

Ovarian – 44 percent (95% CI 36 to 53 percent)

BRCA2 mutation carriers

Breast – 69 percent (95% CI 61 to 77 percent)

Ovarian – 17 percent (95% CI 11 to 25 percent)

Additionally, breast cancer incidence was noted to rise in early adulthood until 30 to 40 years for BRCA1 carriers and until 40 to 50 years for BRCA2 carriers, after which it plateaued at 20 to 30 per 1000 person-years until age 80.

The data from the prospective cohort study above represent cumulative cancer risk until age 80, which is the expected life expectancy of most healthy women in the United States. Note that previous key studies have generally estimated risk only to age 70 [5,29,30,32]. These data for breast and ovarian cancer, as well as for other cancers in BRCA1/2 carriers, are summarized in the table (table 1).

As these figures demonstrate, the lifetime risk of breast cancer is generally somewhat higher with BRCA1 than BRCA2 mutations [29]. BRCA1 carriers also have earlier-onset disease, particularly before age 50 [33-35]. The mean age of diagnosis of breast cancer is younger for BRCA1 than BRCA2 mutation carriers (43 versus 47 years in one study) [33]. BRCA1 carriers are more likely to develop a triple-negative breast cancer than BRCA2 carriers or those who are BRCA1/2 mutation negative [36,37].

Ductal carcinoma in situ (DCIS) is also considered a component of the BRCA1/2 clinical spectrum [38-41]. DCIS occurs at an earlier age in BRCA1/2 carriers than in noncarriers. For example, in one study, BRCA1/2 carriers with DCIS were more than 12 years younger than those with DCIS in the general population [38].

Male breast cancer — Male BRCA1/2 carriers have increased susceptibility to breast cancer, and the risk is higher with BRCA2 versus BRCA1 mutations [42,43]. In men with a BRCA2 gene mutation, one study found the lifetime risk of breast cancer to age 80 to be approximately 3.8 percent as compared with men with a BRCA1 mutation, whose risk is approximately 0.4 percent [44]. Other studies have reported the lifetime risks in BRCA2 carriers to be approximately 7 to 8 percent [42,43]. The lifetime risk of breast cancer for men in the general population is approximately 0.1 percent [45]. In a study including 97 male BRCA1/2 mutation carriers with breast cancer, the median age at diagnosis was 64 (range, 24 to 87), and three men developed contralateral cancers [42]. (See "Breast cancer in men".)

Other gynecologic malignancies

Fallopian tube – At least 50 percent of serous cancers diagnosed in BRCA1/2 carriers are of distal fallopian tube origin [46-48]. The lifetime risk of developing fallopian tube carcinoma in BRCA1/2 mutation carriers is estimated to be 0.6 percent [48], while the general population risk is 0.2 percent [49]. This risk may be an underestimate, given that many high-grade serous ovarian cancers appear to originate in the fallopian tubes [50,51]. Of note, the approach to treatment for ovarian versus fallopian tube carcinomas is identical. In addition, salpingectomy is recommended for all carriers who undergo risk-reducing bilateral salpingo-oophorectomy (rrBSO). (See 'Bilateral salpingo-oophorectomy' below.)

In a retrospective review of 108 women with fallopian tube carcinoma, 33 (30.6 percent) carried a pathogenic mutation (BRCA1, 23 women; BRCA2, 10 women) [52]. The highest frequencies of pathogenic BRCA1/2 variants were identified in women of Ashkenazi Jewish ancestry compared with women of other ancestries (55.6 versus 26.4 percent). In addition, carriers were more likely to be diagnosed before 60 years of age compared with women who did not carry a mutation (40.3 versus 17.4 percent).

Primary peritoneal – The lifetime risk of developing peritoneal cancer in BRCA1/2 carriers is estimated to be 1.3 percent. This estimate was derived from a retrospective review of 22 women of Ashkenazi Jewish ancestry affected with primary peritoneal carcinoma [48]. One of the three germline BRCA1/2 founder mutations was identified in nine women (41 percent). The true incidence of primary peritoneal cancer is difficult to determine, and estimates are limited by the lack of precision in the assignment of site of origin for high-grade, metastatic, serous carcinomas at initial presentation. It is possible, for example, that some cancers classified as "primary peritoneal cancers" may have, in fact, been fallopian tube cancers that were missed.

Endometrial – There are conflicting data on the risk of developing endometrial cancer in patients with pathogenic BRCA mutations [53-57]. One prospective study of 4456 BRCA1/2 carriers found a small increased risk of endometrial cancer for both BRCA1 (standardized incidence ratio [SIR] 1.91, 95% CI 1.06-3.19) and BRCA2 (1.75, 95% CI 0.55-4.23) carriers, although the absolute risk was low [55]. The increased risk was found to be mostly attributable to tamoxifen use; the SIR for women who used tamoxifen was 4.14 (95% CI 1.92-7.87) and 1.67 for women who did not (95% CI 0.81-3.07). The ten-year cumulative risk among the women treated with tamoxifen was 2 percent. Similarly, a second prospective study found an increase in endometrial cancer risk (SIR of 3.5 among BRCA1 carriers and 1.7 among BRCA2 carriers), which persisted in an analysis that accounted for patients being on endocrine therapy [58].

However, two large, prospective studies of BRCA1/2 carriers found no increased risk of endometrial cancer after rrBSO, and results were unaffected by tamoxifen use [56,57]. Thus, at present, hysterectomy at the time of rrBSO is not recommended solely for prevention. (See 'Role for other prophylactic surgery?' below.)

Uterine papillary serous – Several studies have suggested that uterine papillary serous or serous-like carcinoma (UPSC) is part of the BRCA1/2 tumor spectrum, but the overall risk is very low. For example, one study of an unselected population of 151 women with UPSC found that seven women (4.6 percent) had mutations in BRCA1, TP53, and CHEK2 [59]. Two percent of the subjects had a BRCA1 mutation, which is higher than expected but still quite low.

In a subsequent study of 1083 BRCA1/2 carriers undergoing rrBSO without hysterectomy, women with a BRCA1 mutation experienced an increased incidence of serous endometrial cancer compared with what has historically been observed in the general population, though the absolute risk was still small (4 cases observed among 630 women with BRCA1 mutations followed for a median of 5.1 years; observed-to-expected ratio 22.2, 95% CI 6.1-56.9) [56]. Women with BRCA2 mutations did not experience a significantly higher incidence than the general population (1 case per 456 women; observed-to-expected ratio 6.4, 95% CI 0.2-35.5).

Pancreas — Many studies have confirmed an increased risk of pancreatic cancer in BRCA1/2 mutation carriers [53,60-62]. The reported lifetime risk of pancreatic cancer to age 80 ranges between 1 and 3 percent for BRCA1 carriers, and is slightly higher for BRCA2 carriers, ranging between 2 and 5 percent [44,53,61].

In one study of 41 families with a known BRCA1/2 mutation, the median age at diagnosis of pancreatic cancer was reported to be 59 (range, 45 to 80) in male and 68 (range, 38 to 87) in female BRCA1 carriers, respectively. The median age at diagnosis of pancreatic cancer in BRCA2 carriers was reported as 67 (range, 39 to 78) in men and 59 (range, 46 to 81) in women [63]. As in sporadic cases, survival in BRCA1/2 mutation carriers is poor [64].

Prostate — Studies have shown that men with a BRCA2 mutation have between a five- and ninefold increased risk of prostate cancer, estimated to translate to a risk of about 30 percent [44,60,65]. The risk of prostate cancer in men with a BRCA1 mutation is less clear and data are mixed; for example, one study found the risk to be elevated by about 3.75-fold (translating to a risk of about 9 percent by age 65) [66], while a separate study did not find an increase among BRCA1 carriers [44].

However, risk scores based on polygenic (single nucleotide polymorphism) testing, which are not routinely integrated into clinical care, have also been used to predict risks in BRCA1/2 carriers [67]. Using the 50th percentile cutoff of polygenic risk score (PRS) distributions, the lifetime risk for BRCA1 carriers is on the order of 15 to 20 percent, and 30 to 40 percent for BRCA2 carriers. The absolute risk range to age 80 across the PRS distribution was wide: 7 to 26 percent for BRCA1 and 19 to 61 percent for BRCA2. Note that the general population risk of prostate cancer varies by race and ethnicity; therefore, absolute risk estimates are not necessarily reflective of these differences.

Given that a significant proportion of men in the general population have prostate cancer, many cases of which are found at autopsy, the baseline population risk is difficult to estimate. Determining the absolute risk in BRCA1/2 carriers is similarly hard to pinpoint, and there is concern about whether the increased risks observed in carriers are the result of over-diagnosis. However, several studies have shown that prostate cancer in BRCA1/2 carriers is more aggressive and has poorer survival outcomes relative to the general population [68-71].

Other solid tumors — Aside from the risks of breast, ovarian, prostate, and pancreatic cancers, risks of cancer at other sites in BRCA1/2 carriers are based on low numbers of affected individuals, often with unconfirmed diagnoses, and findings have not been widely replicated. Thus, the clinical relevance of these data is uncertain, and the role of cancer screening and risk reduction options is undefined.

Colorectal – Data regarding risk of colorectal cancer (CRC) in BRCA1/2 carriers are inconsistent. Although some studies have reported an increased risk of CRC among BRCA1 mutation carriers, other population-based series have not confirmed such an association. Further details are found elsewhere. (See "Epidemiology and risk factors for colorectal cancer", section on 'Are patients with HBOC syndrome at risk?'.)

Melanoma and non-melanomatous skin cancers – An association between BRCA2 pathogenic variants and cutaneous melanoma has been described, but the risk is not well characterized [60,72,73]. No clear association with BRCA1 has been noted.

The risk of uveal melanoma, which is very rare, is increased in BRCA2 carriers. For example, in a study of 222 BRCA1/2 families, two BRCA2 carriers developed this malignancy, which translated to a relative risk of 99.4 (95% CI 11.1-359.8) [74]. Because of the wide confidence interval, translation to an absolute risk is not possible. The risk of uveal melanoma in BRCA1 carriers is not well described [75].

Studies have not provided a definitive answer about whether the risk of non-melanoma skin cancer is elevated in BRCA1/2 carriers [73].

Stomach and biliary – An increased risk of biliary and gastric cancers in BRCA1 and 2 carriers and esophageal cancers in BRCA2 carriers has been described [44,76]. In a large international study the absolute risks of these cancers were low, for example in BRCA2 carriers the risk of stomach cancer was 3.5 percent up to age 80 years [44]. In a study from Japan, however, the risk of gastric cancer in BRCA1 and BRCA2 carriers was about 19 to 20 percent up to age 85 [76]. This discrepancy may in part reflect the high risk of this malignancy in East Asian countries.

MANAGEMENT OF FEMALE BRCA1/2 CARRIERS WITHOUT CANCER — For women without a personal history of cancer in whom a BRCA1/2 pathogenic variant is identified, national guidelines recommend risk-reducing bilateral salpingo-oophorectomy (rrBSO) once childbearing is complete, and between the ages of 35 and 40. As BRCA2 carriers tend to develop ovarian cancer 8 to 10 years later than BRCA1 carriers, delaying rrBSO until age 40 to 45 in BRCA2 carriers can be considered. Studies show that rrBSO significantly reduces the risk of ovarian cancer and improves survival. (See 'Bilateral salpingo-oophorectomy' below.)

Specific recommendations for breast cancer risk management include intensive screening as well as consideration of hormonal and surgical forms of risk reduction. In addition to bilateral mastectomy, premenopausal women should be informed that obtaining rrBSO by the recommended age may reduce the risk of breast cancer. (See 'Risk-reducing surgery' below.)

Clinical decision-making around which strategies to pursue for cancer risk reduction (ie, surveillance, risk-reducing surgery, and/or chemoprevention) involves a trade-off between life expectancy and quality of life. Several studies have suggested that decision aids or data from models may help patients choose among different options [77-81], and most use decision analysis and the concept of time tradeoffs (ie, years of life saved by one strategy as compared with another). While useful from a clinical research standpoint, they are not often employed in clinical practice.

Counseling regarding modifiable lifestyle choices — Although several studies have reported associations between specific risk factors and breast/ovarian cancer risks in BRCA1/2 carriers, a rigorous meta-analysis of 44 nonoverlapping studies assessing several hormonal and several exogenous risk factors found that the only factor to be clearly associated with breast cancer risk was age at first live birth [82].

Specifically, a meta-analysis of two cohort studies found that later age at first live birth for BRCA1 carriers may be protective against breast cancer [82]. A reduced effect size (ES) was found when comparing women who had delivered their first child at age 30 years and older with women at age 25 to 29 years at the time of delivery (ES 0.69, 95% CI 0.48-99). However, there was no impact on age at first birth noted for BRCA2 carriers (ES 1.02, 95% CI 0.64-1.63).

With respect to other risk factors, results from the meta-analysis showed that the following may have possible associations with cancer risks:

Increased breast cancer risk:

Oral contraceptives (theoretic, both for BRCA1 and BRCA2)

Smoking (BRCA2)

Decreased breast cancer risk:

Breastfeeding (BRCA1)

Late age at menarche (BRCA1)

Decreased ovarian cancer risk:

Breastfeeding (BRCA1)

Tubal ligation (BRCA1)

Oral contraceptive use (BRCA1 and BRCA2)

In summary, while data are limited in BRCA1/2 carriers, general information about specific risk factors may be useful for counseling individual women about their risk and modifiable lifestyle choices.

Risk-reducing surgery — For women without a personal history of cancer who have a BRCA1/2 pathogenic variant, prophylactic (or preventative) surgery reduces the risk of developing cancer. However, risk-reducing surgery does not completely eliminate the risk of developing cancer, as residual risks remain after mastectomy and bilateral salpingo-oophorectomy. While prophylactic surgery is effective in cancer risk reduction, women should be counseled preoperatively about the potential morbidity of such procedures, and the possibility that surgery may affect libido, sexual functioning, and body image. Oophorectomy in premenopausal women can be associated with increased risks for bone and heart disease, and raises concerns about how to optimally manage surgical menopause and hormone therapy. These procedures are discussed further below.

Mastectomy — We agree with the National Comprehensive Cancer Network and recommend that BRCA1/2 carriers be offered risk-reducing bilateral mastectomy [83]. However, the decision about whether or not to undergo such surgery is based on personal preference, given that effective screening is available. (See 'Breast cancer screening' below.)

In both retrospective and prospective observational studies, risk-reducing or prophylactic bilateral mastectomy decreases the incidence of breast cancer by 90 percent or more in patients at risk of hereditary breast cancer, with most studies focusing on BRCA1/2 mutation carriers [84-93]. As an example, in one large, prospective, multi-institutional international study that included over 2400 women with a known BRCA1/2 mutation, no patients developed breast cancer following mastectomy (0 of 247 women), whereas 98 of 1372 women (7 percent) who did not undergo mastectomy did [90].

Patients who opt to proceed with mastectomy are most often undergoing skin-sparing mastectomy with or without preservation of the nipple-areolar complex followed by immediate breast reconstruction. Nipple-areolar-sparing mastectomy in particular provides superior cosmetic results, with low recurrence risks [94,95]. As an example, in a multi-institution review of 346 BRCA1/2 carriers undergoing either bilateral mastectomy or contralateral mastectomy with nipple-areolar sparing there were no cases of breast cancer, whereas, based on models, 22 would have been expected [94]. Although the follow-up was relatively brief (median, 34 months), in experienced hands, this procedure is considered to be an acceptable option for risk reduction. (See "Mastectomy", section on 'Skin-sparing mastectomy' and "Mastectomy", section on 'Nipple-areolar-sparing mastectomy'.)

Bilateral salpingo-oophorectomy — For BRCA1/2 carriers, rrBSO is recommended for women who have completed childbearing, and should be performed by age 35 to 40 or individualized based on age of onset of ovarian cancer in the family [83,96-99]. In BRCA2 carriers, one can consider delaying this procedure until age 40 to 45. Whenever possible, patients should be referred to a gynecologic oncologist for a discussion about this surgery, and specific protocols have been recommended for pathologic review and follow-up of abnormal findings [83]. Salpingectomy and delayed oophorectomy, as a means of preserving fertility during childbearing years, has also been evaluated and is discussed elsewhere. (See "Risk-reducing salpingo-oophorectomy in patients at high risk of epithelial ovarian and fallopian tube cancer", section on 'BSO versus salpingectomy alone'.)

Improvements in ovarian cancer risks and mortality – rrBSO not only decreases the risk of ovarian cancer in BRCA1/2 mutation carriers, but also decreases mortality [90,97,98]. In a 2014 meta-analysis of three prospective studies of rrBSO in BRCA1/2 carriers, the procedure was associated with an 80-percent reduction in ovarian cancer (risk ratio [RR] 0.19, 95% CI 0.13-0.27) and a 68-percent reduction in all-cause mortality (RR 0.32, 95% CI 0.27-0.38) [100].

Continued risk of peritoneal carcinoma after rrBSO – Peritoneal carcinoma should be considered a phenotypic variant of ovarian cancer. Women who undergo rrBSO remain at risk for developing this cancer [101-104], which is much less common than ovarian cancer but associated with high mortality, similar to that of stage III epithelial ovarian cancer [105,106]. A review of five studies including 846 patients with BRCA1/2 mutations concluded the risk of peritoneal cancer after rrBSO was 1.7 percent (range, 0.5 to 10.7 percent) [107]. Subsequently, a large prospective study including 1045 patients estimated a 4.3 percent cumulative incidence of peritoneal carcinoma in BRCA1/2 mutation carriers at 20 years after rrBSO [108]. The risk appears to be highest in BRCA1 carriers, although an association with BRCA2 cannot be excluded; peritoneal carcinomas have been reported in a few women with BRCA2 mutations [108-110]. Furthermore, risk appears to be higher among patients with serous tubal intraepithelial carcinoma (STIC) found at the time rrBSO (hazard ratio [HR] 33.9), translating to a 10-year risk of 28 percent, versus 0.9 percent for those without STIC at the time of surgery [104]. Malignancy found at the time of surgery is discussed in more detail elsewhere. (See "Risk-reducing salpingo-oophorectomy in patients at high risk of epithelial ovarian and fallopian tube cancer", section on 'Occult malignancy on pathology'.)

Effect on breast cancer risk – Although previous data suggest that rrBSO may be beneficial for reduction of breast cancer risk, other data have called this into question, particularly for BRCA1 carriers [111]:

In the 2010 prospective study described above [90], rrBSO was also found to decrease breast cancer risk in BRCA1 (HR 0.63, 95% CI 0.41-0.96) and BRCA2 carriers (HR 0.36, 95% CI 0.16-0.82).

However, in an analysis of a nationwide cohort of over 800 BRCA1/2 carriers with no prior personal history of cancer, the HR for breast cancer following rrBSO was 1.09 (95% CI 0.67-1.77) [98]. This analysis aimed to minimize biases due to selection, lead time, and short follow-up. The authors noted that rrBSO, when performed prior to menopause, may have a slight protective effect on breast cancer risk.

Finally, a study of 3722 BRCA1/2 carriers unaffected by cancer with 5.6 years' follow-up found that rrBSO had a nonsignificant trend to a reduced risk of breast cancer in BRCA2 carriers (HR 0.65, 95% CI 0.37-1.16), but not BRCA1 carriers (HR 0.96, 95% CI 10.73-1.26). In analyses evaluating effect of rrBSO when stratified by age, reduction in breast cancer risk in BRCA2 mutation carriers diagnosed prior to age 50 years was statistically significant (age-adjusted HR 0.18, 95% CI 0.05-0.63), but was not in BRCA1 carriers (age-adjusted HR 0.79, 95% CI 0.55-1.13) [112].

Thus, we concur with the recommendation for BRCA1/2 carriers to undergo rrBSO with the primary aim to decrease the risk of and mortality attributable to ovarian and fallopian tube cancer. Based on available data, we feel that rrBSO also reduces the risk of breast cancer in BRCA2 carriers prior to age 50, but not in BRCA1 carriers. (See "Risk-reducing salpingo-oophorectomy in patients at high risk of epithelial ovarian and fallopian tube cancer".)

Women who undergo rrBSO will likely experience side effects of surgically induced menopause. Discussion of hormone therapy (HT) for management of menopausal symptoms is found below. These women are also at an increased risk of developing osteoporosis. They should be counseled about osteoporosis prevention and screening. (See 'Hormone therapy' below and "Screening for osteoporosis in postmenopausal women and men" and "Overview of the management of low bone mass and osteoporosis in postmenopausal women", section on 'Lifestyle measures to reduce bone loss'.)

Role for other prophylactic surgery? — The only proven risk-reducing procedure for ovarian cancer in BRCA1/2 mutation carriers is rrBSO. We do not routinely recommend hysterectomy at the time of rrBSO for prevention purposes. Hysterectomy may be indicated for other reasons or to allow the use of unopposed estrogen replacement therapy. Of note, there are no national guidelines that recommend routine hysterectomy in BRCA1/2 mutation carriers. While studies have reported a possible small, excess risk of uterine cancers in mutation carriers [56,58,59,113], the absolute risk is low, and it is not clear that the benefits associated with hysterectomy are sufficiently large enough to warrant the risks associated with surgery. While hysterectomy is not indicated to reduce cancer risk, some carriers may, however, choose to undergo this surgery at the time of rrBSO in order to enable them to take unopposed estrogen HT without increasing their risk of endometrial cancer. It is therefore critical to have at least an initial discussion about these options preoperatively. (See 'Hormone therapy' below.)

There is controversy about whether it is appropriate to perform a salpingectomy alone for BRCA1/2 mutation carriers who wish to defer oophorectomy [114,115], based upon a possible fallopian tube origin for some ovarian cancers [116,117]. The Society of Gynecologic Oncology (SGO) Clinical Practice Statement states that salpingectomy may be appropriate and feasible as a strategy for ovarian risk reduction, but further study is needed to determine safety of this practice [114]. However, the statement and a lengthier explication make clear that this procedure does not eliminate the risk of ovarian cancer, and it does not reduce the risk of breast cancer [114,118]. Guidelines from the National Comprehensive Cancer Network thus indicate that salpingectomy alone is not the standard of care [83].

Ongoing randomized controlled trials or prospective clinical trials are evaluating this question but until there are sufficient data from these studies, we do not offer salpingectomy without an oophorectomy for these women. (See "Opportunistic salpingectomy for ovarian, fallopian tube, and peritoneal carcinoma risk reduction".)

Hormone therapy

Clinical approach — Prior to using HT to manage menopausal symptoms in BRCA1/2 carriers who have undergone rrBSO, a shared decision-making process must include counseling women about nonhormonal options and the lack of population-specific data regarding HT.

While the data are somewhat limited, we feel that estrogen-alone HT is a reasonable option for younger mutation carriers [119], particularly those who have undergone risk-reducing mastectomy and hysterectomy. For carriers not undergoing risk-reducing mastectomy, we counsel regarding a theoretically increased breast cancer risk, although data are limited. Nonhormonal options are also an option for menopausal symptoms and osteoporosis prevention and treatment in BRCA carriers who have undergone rrBSO. (See "Menopausal hot flashes", section on 'Nonhormonal pharmacotherapy'.)

Women who plan to take systemic HT after rrBSO should be counseled about the possible risks and benefits of HT and possible ways to mitigate these risks. These include:

Limiting the duration of HT – Some clinicians begin HT after surgery and then make a plan with the patient to taper it within several years. Another approach is to stop HT at 51 years old, the average age of natural menopause.

Concurrent hysterectomy – To enable use of unopposed estrogen for management of menopausal symptoms without incurring a risk of endometrial cancer, some women choose to undergo concomitant hysterectomy at the time of rrBSO. However, hysterectomy is not otherwise routinely indicated, as discussed above. (See 'Role for other prophylactic surgery?' above.)

The rationale for possible hysterectomy in this setting is as follows. Estrogen therapy alone appears to incur a lower risk of breast cancer than estrogen-progestin therapy. However, unopposed estrogen therapy in a woman with a uterus leads to endometrial cancer. A potential alternative is an ultra-low-dose transdermal estrogen formulation, with which progestin opposition is required only periodically (eg, every six to 12 months). (See "Preparations for menopausal hormone therapy", section on 'Transdermal estrogen'.)

Prophylactic mastectomy. (See 'Mastectomy' above.)

Systemic hormone therapy — Our approach to HT in patients who have undergone rrBSO is discussed above. (See 'Clinical approach' above.)

There are no high-quality data in this setting, but overall, studies suggest that the risk of breast cancer is not increased in younger BRCA carriers (≤45 years) taking HT after rrBSO; among carriers older than 45 years, studies are conflicting, but there may be an increased risk of breast cancer, potentially related to time of exposure to HT around the natural age of menopause [90,120-122]. Examples of available data are as follows:

In a prospective cohort study of 872 BRCA1 mutation carriers who had undergone oophorectomy, HT was not associated with an increased risk of breast cancer at a median follow-up of 7.6 years (HR 0.97, 95% CI 0.62-1.52) [120]. In carriers taking any type of HT, there was no increase in risk of subsequent breast cancer compared with controls. However, in those taking estrogen plus progesterone HT, the 10-year actuarial risk of breast cancer was 22 percent, as compared with 12 percent in those taking estrogen alone. There were no statistically significant differences based on age.

In a retrospective cohort study of 306 BRCA1/2 mutation carriers who had undergone rrBSO, 36 patients were diagnosed with breast cancer at a median follow-up of 7.3 years, 13.5 percent in the HT group and 10.3 percent in the non-HT group (OR 1.4, 95% CI 0.7-2.7) [123]. In women who were ≤45 years at the time of rrBSO, HT did not affect breast cancer rates; however, those older than 45 years who took HT had higher rates of breast cancer than those who did not take HT (21 versus 8 percent; OR 3.4, 95% CI 1.2-9.8).

There is a substantial body of evidence among non-BRCA1/2 carriers regarding the association of exogenous HT with breast cancer, predominantly hormone receptor-positive breast cancer. Representative data are as follows:

The most influential data come from the Women's Health Initiative trial, which showed that the use of combined estrogen-progestin HT resulted in a significantly increased risk of breast cancer in postmenopausal women over the age of 50. By contrast, there was no increase in breast cancer risk in women who received unopposed estrogen. The relationship between HT and breast cancer is discussed in detail separately. (See "Menopausal hormone therapy and the risk of breast cancer".)

In breast cancer survivors, data from randomized trials show a significant increase in rates of breast cancer recurrence with the use of HT; this risk appears to be present for both estrogen-progestin and estrogen-alone therapy, although data regarding specific regimens are limited [124]. (See "Menopausal hormone therapy and the risk of breast cancer", section on 'Personal history of breast cancer'.)

However, the generalizability of these data to premenopausal women who undergo rrBSO is unclear. These women differ from the postmenopausal population studied in the Women's Health Initiative trial: they are likely to be younger than age 51 and have had surgical rather than natural menopause. Additionally, although BRCA carriers are at a high risk of breast cancer, they cannot be equated with women who have been diagnosed with breast cancer.

Vaginal estrogen therapy — For carriers with symptomatic vaginal atrophy, nonhormonal options may be used as first-line therapy. However, we have a low threshold to use vaginal estrogen to treat persistent symptoms. (See "Genitourinary syndrome of menopause (vulvovaginal atrophy): Treatment".)

Vaginal estrogen therapy is the most effective treatment for vaginal atrophy. It alleviates symptoms of dyspareunia and vaginal dryness, and may also result in decreased incidence of urinary tract infections and overactive bladder symptoms. Low-dose vaginal estrogen therapy for treatment of local symptoms typically does not raise the serum estrogen concentration above the average level following natural menopause. There are no data regarding the risk of breast cancer in high-risk women treated with vaginal estrogen therapy.

Cancer surveillance — For female BRCA1/2 carriers who do not wish to pursue (or would rather delay) surgical risk reduction, breast cancer surveillance should be offered, and ovarian cancer screening may be performed [83]. While breast and ovarian (including fallopian tube and peritoneal) cancers present the greatest risk, carriers have elevated risks for other cancers, including prostate cancer and pancreatic cancer. (See "Overview of hereditary breast and ovarian cancer syndromes".)

Women with BRCA1/2 pathogenic variants should be educated regarding signs and symptoms of breast and ovarian cancer as appropriate. (See "Clinical manifestations, differential diagnosis, and clinical evaluation of a palpable breast mass" and "Epithelial carcinoma of the ovary, fallopian tube, and peritoneum: Clinical features and diagnosis" and "Breast cancer in men" and "Breast cancer in men", section on 'Presentation'.)

Breast cancer screening — The following screening strategy is recommended by expert groups for women with BRCA1/2 pathogenic variants who have not undergone risk-reducing surgery, and should be individualized as needed [83,96,125]:

Beginning at age 18, self-breast exams performed periodically may facilitate awareness of changes, and clinical breast examination should be performed every 6 to 12 months beginning at age 25.

Magnetic resonance imaging (MRI) for breast cancer screening is recommended annually beginning at age 25, or earlier depending on the earliest age of breast cancer in the family [83,126].

Mammography with consideration of tomosynthesis should begin at age 30 or be individualized if the earliest age of onset in the family is under age 25. Breast MRIs and mammography may be staggered by 6 months.

However, the sensitivity of mammography for detecting breast cancer in mutation carriers appears to be lower than in other high-risk women, which may be due to [83,96,127-129]:

Higher breast density [129].

Differences in morphologic features (eg, less spiculation due to lack of tumor-surrounding fibrosis) [128].

The frequent development of interval malignancies [130].

The addition of breast MRI to the breast cancer surveillance strategy in high-risk women increases breast cancer detection rates, increases the number of patients diagnosed at an earlier stage of disease, is cost effective (image 1) [79,81,131-133], and is supported in multiple guidelines [83,125,134]. However, the mortality impact of including breast MRI in the surveillance strategy is not clear. Representative data are as follows:

In a systematic review of 11 nonrandomized studies in which high-risk women were screened by both MRI and mammography, MRI was more sensitive but less specific than mammography, but the combination of the two modalities was both more sensitive and specific compared with mammography alone [135]. For example, the sensitivity and specificity of detecting a Breast Imaging Reporting and Data System (BI-RAD) ≥4 lesion were 32 and 99 percent, respectively, with mammography; 75 and 96 percent, respectively, with MRI; and 84 and 99 percent, respectively, with combined-modality therapy.

In a prospective study in which women with a high risk for breast cancer were screened with annual mammography and MRI, and were followed for a median of five years if they developed breast cancer, the overall survival at six years in BRCA1/2 carriers with invasive cancer was 93 percent (95% CI 79.0-97.6) [131]. While these data are promising, the lack of a control group makes it hard to conclude that MRI was associated with a survival benefit.

In a prospective cohort study of 1275 BRCA1/2 carriers followed for a mean of 3.2 years, the addition of MRI to mammography did not increase the number of women diagnosed with breast cancer, but those screened with MRI were more likely to be diagnosed with ductal carcinoma in situ (DCIS) or stage I disease (13.8 versus 7.2 percent, respectively) as opposed to stage II to IV disease (1.9 versus 6.6 percent, respectively) [132]

While the risk of radiation-associated breast cancer from breast imaging for the average-risk patient is believed to be small or nonexistent, women at high genetic risk may be more susceptible to radiation-induced carcinogenesis because of the role of BRCA1/2 proteins in DNA repair. In a retrospective study of 1993 carriers of BRCA1/2 mutations, any exposure of diagnostic radiation before age 30 was associated with an increased risk of breast cancer (HR 1.90, 95% CI 1.20-3.00), with a dose-response pattern. By contrast, in a report based upon a questionnaire sent to known BRCA1/2 mutation carriers, exposure to mammographic screening (mean age at first screening mammogram, 35 years of age) was not associated with an increased risk of breast cancer when adjusted for parity, oral contraceptive use, family history, and ethnicity. Further research is needed, and current recommendations by most groups support screening these women with a combination of mammography and MRI scanning rather than breast MRI exclusively [136-139].

Data from large studies evaluating the role of breast ultrasound in addition to other imaging modalities have not demonstrated any additional benefit of this procedure [140-142]. Thus, we do not routinely recommend it to mutation carriers who are already undergoing MRI and mammogram.

Ovarian cancer screening — For carriers who are of the age range of recommended rrBSO and opt not to pursue it, we offer ovarian cancer screening. This consists of concurrent transvaginal ultrasound (preferably day 1 to 10 of menstrual cycle) and cancer antigen (CA) 125 (best performed after day 5 of menstrual cycle) every six months beginning at age 30 or 5 to 10 years before the earliest age of first diagnosis of ovarian cancer in the family according to previous guidelines from the National Comprehensive Cancer Network [143]. However, we acknowledge that 2023 guidelines do not discuss this strategy and there is a lack of high-quality data to inform these recommendations; as such, some patients may reasonably prefer not to undergo screening [144].

Screening for other cancers — There is no consensus regarding screening for melanoma or pancreatic cancer; however, recommendations may be based on an individual's family history. Possible recommendations in carriers include full-body skin exams and pancreatic cancer screening, for those with affected relatives [83]. However, there was no consensus about what age to initiate screening, how often to repeat it, and how to follow up after abnormal findings. We generally recommend that BRCA2 carriers obtain full-body skin exams as a precaution, and follow the International Cancer of the Pancreas Screening Consortium guidelines for screening for pancreatic cancer, as detailed elsewhere. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients", section on 'Pancreatic cancer screening'.)

With respect to colon cancer screening, guidelines for management in BRCA1/2 carriers do not differ from those in the general population. However, if an individual has a family history of colon cancer or a prior history of adenomas or other conditions that may increase risk, more aggressive screening may be recommended [83].

Chemoprevention

Tamoxifen — For female BRCA2 carriers who opt against mastectomies, we offer tamoxifen or aromatase inhibitors (AIs) for risk reduction for women. However, based on the patient's age and general health, the option of risk-reducing mastectomy should be rediscussed periodically with patients, as medical chemoprevention is less effective than surgery. (See "Selective estrogen receptor modulators and aromatase inhibitors for breast cancer prevention", section on 'BRCA carriers'.)

Only limited data are available regarding the preventive benefit of tamoxifen in BRCA1/2 mutation carriers. Evidence for the benefit of tamoxifen in women who have never had a diagnosis of breast cancer comes from a subset analysis of the National Surgical Adjuvant Breast and Bowel Project (NSABP) Breast Cancer Prevention trial (P-1 trial). Tamoxifen reduced breast cancer risk by 62 percent in BRCA2 carriers (relative risk [RR] 0.38, 95% CI 0.06-1.56), but not in BRCA1 carriers (RR 1.67, 95% CI 0.32-10.07) [145]. However, this analysis is limited by the small number of mutation carriers (of the 288 women in the study who developed breast cancer, only 8 had BRCA1 pathogenic variants and 11 had BRCA2 pathogenic variants) [146]. There are no data addressing the preventive benefit of raloxifene or an AI in patients with BRCA1/2 mutations. However, in large chemoprevention studies of postmenopausal women at increased risk for breast cancer, both raloxifene and AIs have demonstrated reductions in the risk of breast cancer [147-149].

A differential effect of tamoxifen in BRCA2 as compared with BRCA1 mutation carriers may be attributed to estrogen receptor (ER) status of BRCA1- and BRCA2-associated tumors. Tamoxifen might be expected to have an impact only against ER-positive tumors, and BRCA2-associated tumors have a greater likelihood than BRCA1-associated tumors of being ER positive.

Oral contraceptives — We suggest that all carriers undergo rrBSO at an age before which they are at the highest risk for ovarian cancer, and therefore oral contraceptives as chemoprevention agents are not indicated. For patients who have taken oral contraceptives (or who have opted against rrBSO), we discuss that evidence supports a reduced risk of ovarian cancer in carriers who take oral contraceptive pills, although a theoretic risk of increased breast cancer exists. However, this has not been clearly supported in the literature.

A meta-analysis of 18 comparative, retrospective studies of oral contraceptive use in BRCA1/2 mutation carriers included 1503 cases of ovarian cancer and 2855 cases of breast cancer [150]. Ever-use of oral contraceptives in BRCA1/2 mutation carriers was associated with a reduced risk of ovarian cancer (RR 0.50, 95% CI 0.33-0.75); this effect was found to a similar degree in both BRCA1 and BRCA2 mutation carriers. The protective effect increased with longer duration of use. The largest study in the meta-analysis, a case-control study that included 798 women with ovarian cancer, found a 5 percent decrease in risk of ovarian cancer per year of oral contraceptive use [151]. In addition, oral contraceptive use appears to be associated with a decreased risk of fallopian tube cancer in the general population [152].

There has been concern that oral contraceptives may increase the risk of breast cancer in mutation carriers. In the meta-analysis described above, there was no evidence of a significantly increased breast cancer risk in oral contraceptive users overall, for users of current formulations of oral contraceptives, or in the first 10 years after cessation of use [150]. (See 'Chemoprevention' above.)

One limitation of these studies is that all the data are for use of oral contraceptives. Other formulations of estrogen-progestin contraceptives (patch, vaginal ring) have not been studied in this clinical context; thus, it is unknown whether they provide similar ovarian cancer prevention.

Chemoprevention of ovarian cancer with agents other than estrogen-progestin contraceptives (eg, vitamin D) is under investigation. Protective factors against ovarian cancer in the general population are discussed in detail separately. (See "Epithelial carcinoma of the ovary, fallopian tube, and peritoneum: Incidence and risk factors", section on 'Protective factors'.)

Fertility — Evidence is mixed regarding whether BRCA carriers have decreased fertility. While several studies have suggested a lower serum anti-Müllerian hormone (AMH) in BRCA carriers relative to controls [153-155], others have not confirmed this finding [154,156]. In the largest available analysis (250 BRCA carriers and 578 controls), it was suggested that female BRCA1 carriers had an approximately 33 percent lower AMH level relative to controls, although levels in BRCA2 carriers were not diminished [157]. This finding may represent decreased ovarian reserve, and therefore fertility counseling may be appropriate for BRCA1 carriers, if decisions regarding chemotherapy or delayed childbearing are being considered. Discussion of the approach to infertility is discussed elsewhere. (See "Overview of infertility".)

MANAGEMENT OF MALE BRCA1/2 CARRIERS WITHOUT CANCER — For men, screening for breast and prostate cancers is generally recommended.

Cancer surveillance — There are no proven risk-reducing surgical options for male mutation carriers. Therefore, the following screening strategy is recommended for men (with BRCA1/2 mutations) (see "Breast cancer in men"):

Monthly breast self-examination starting at age 35.

Clinical breast examination every 12 months starting at age 35.

Mammography – Guidelines now recommend consideration of annual mammogram, particularly for BRCA2 carriers, beginning at age 50 or 10 years before the earliest known male breast cancer in the family [83].

Prostate cancer screening starting at age 40 for BRCA2 carriers and consideration of prostate screening for BRCA1 carriers at age 40 [83]. Other guidelines also suggest that age at diagnosis of any affected family members should be factored into screening discussions [158]. (See "Screening for prostate cancer".)

A potential benefit of prostate-specific antigen screening is that BRCA1/2 status may be a factor in deciding whether men with early-stage/localized prostate cancer choose surgery versus active surveillance [158]. Given the potentially aggressive nature of BRCA1/2-associated prostate cancers, the former option may be preferred. (See "Localized prostate cancer: Risk stratification and choice of initial treatment".)

The approach to screening for other cancers is identical to that adopted for female BRCA1/2 carriers. (See 'Screening for other cancers' above.)

Our approach to cancer surveillance is generally the same for male BRCA1 and BRCA2 carriers (with one possible exception being prostate cancer). Although results from a cohort study of 6900 male BRCA1/2 carriers suggested differences in the cancer spectrum between male BRCA2 versus BRCA1 carriers (eg higher risks of breast, prostate, and pancreatic cancer, but lower risk of colorectal cancer) [159], further data are needed to confirm these differences.

Role for chemoprevention? — Although tamoxifen is used to treat some men with hormone receptor-positive breast cancer, it is not recommended for use as chemoprevention in BRCA1/2 carriers, whose overall risk is low. (See "Breast cancer in men".)

Given the high risk of prostate cancer in male BRCA1/2 carriers, especially in BRCA2 carriers, the question has been raised about whether they may be good candidates for chemoprevention with agents such as 5-alpha-reductase inhibitors. However, there are no data regarding the effectiveness of chemoprevention in male carriers. Information about prostate cancer chemoprevention is discussed separately. (See "Chemoprevention strategies in prostate cancer".)

REPRODUCTIVE COUNSELING — Pathogenic variants in many breast cancer genes, including BRCA, are inherited in an autosomal-dominant pattern, meaning that there is a 50 percent chance that children of BRCA1/2 carriers will have inherited the cancer-predisposition variant. Reproductive counseling of BRCA1/2 carriers includes education about prenatal diagnosis and assisted reproduction [143]. One option is preimplantation genetic diagnosis, which is used to analyze embryos (obtained by in vitro fertilization) genetically before their transfer into the uterus [160]. (See "Preimplantation genetic testing".)

In addition, BRCA2 carriers who plan to have children with a partner who is also at increased risk of carrying a BRCA2 pathogenic variant (eg, owing to personal/family cancer history or to the increased background frequency in individuals of Ashkenazi Jewish descent) are at risk of having offspring who may inherit a rare, recessive syndrome that is characterized by the co-occurrence of brain tumors, Fanconi anemia, and breast cancer [161]. This syndrome occurs in individuals who have two (biallelic) mutations in BRCA2. Biallelic mutations in BRCA1 and other genes may also predispose to Fanconi anemia and other severe disorders; thus, reproductive risks should be discussed with these carriers as well [83,162,163].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Hereditary breast and ovarian cancer".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Genetic testing for breast, ovarian, prostate, and pancreatic cancer (The Basics)")

Beyond the Basics topics (see "Patient education: Genetic testing for hereditary breast, ovarian, prostate, and pancreatic cancer (Beyond the Basics)" and "Patient education: Medications for the prevention of breast cancer (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Introduction – Women with inherited pathogenic variants in breast cancer type 1 and 2 susceptibility genes (BRCA1 and BRCA2; referred in this topic as BRCA1/2) have markedly elevated risks of breast and ovarian cancer. Male carriers have increased risk for breast and prostate cancer. (See 'Introduction' above.)

Breast cancer risk reduction – For women with a BRCA1/2 pathogenic variant, we offer risk-reducing mastectomy. (See 'Mastectomy' above.)

For women with a BRCA1/2 pathogenic variant who do not opt for bilateral mastectomy, we offer surveillance with annual mammography and magnetic resonance imaging (MRI). We initiate MRI at age 25 and mammography at age 30. (See 'Cancer surveillance' above.)

For female BRCA2 carriers who do not opt for risk-reducing mastectomy, we suggest endocrine therapy for chemoprevention (Grade 2C). Although tamoxifen is the only agent that has been studied specifically in BRCA1/2 carriers, we extrapolate from studies demonstrating a protective effect of aromatase inhibitors and raloxifene in other high-risk postmenopausal women, and offer these agents to postmenopausal women who prefer these options based on side-effect profile. (See 'Chemoprevention' above and "Selective estrogen receptor modulators and aromatase inhibitors for breast cancer prevention".)

Epithelial ovarian cancer risk reduction – For women with a BRCA1/2 pathogenic variant, we recommend performing risk-reducing bilateral salpingo-oophorectomy (rrBSO) between age 35 and 40 for BRCA1 carriers and between 40 and 45 for BRCA2 carriers, when childbearing is complete, rather than screening (Grade 1B). Of note, an early age at diagnosis of ovarian cancer in the family may prompt consideration of such surgery at a younger age. The age for rrBSO may be younger based on age of onset within the family. (See 'Bilateral salpingo-oophorectomy' above.)

For carriers who have not undergone rrBSO, we discuss ovarian cancer screening with concurrent transvaginal ultrasound and cancer antigen (CA) 125. The ultrasound is preferably performed on days 1 to 10 of menstrual cycle, while CA 125 is best assessed after day 5 of menstrual cycle. These assessments may be performed every 6 months beginning at age 30 to 35, or 5 to 10 years before the earliest age of first diagnosis of ovarian cancer in the family. However, we acknowledge a lack of high-quality data to inform these recommendations, and patients may reasonably opt to avoid these screening procedures. (See "Screening for ovarian cancer", section on 'Lack of benefit of screening strategies'.)

Screening for other cancers – There is no consensus regarding screening for melanoma or pancreatic cancer; however, recommendations may be based on an individual's family history. Increasingly, pancreatic cancer screening is considered in carriers with first- or second-degree relatives with pancreatic cancer. Additionally, full-body skin exams can be offered, particularly for BRCA2 carriers. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients", section on 'Pancreatic cancer screening'.)

Males with BRCA1/2 pathogenic variants – For men with BRCA1/2 pathogenic variants, cancer surveillance options are limited. We encourage clinical and self-breast examination and also suggest mammography, particularly for BRCA2 carriers. We initiate mammography at age 50 or 10 years prior to the earliest known breast cancer in the family. However, given limited data, some males may opt not to pursue mammography.

We also perform prostate cancer screening, beginning at age 40 to 45. (See 'Cancer surveillance' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Suzanne W Fletcher, MD, who contributed to an earlier version of this topic review.

  1. Couch FJ, Nathanson KL, Offit K. Two decades after BRCA: setting paradigms in personalized cancer care and prevention. Science 2014; 343:1466.
  2. Alsop K, Fereday S, Meldrum C, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol 2012; 30:2654.
  3. Szabo CI, King MC. Population genetics of BRCA1 and BRCA2. Am J Hum Genet 1997; 60:1013.
  4. Ferla R, Calò V, Cascio S, et al. Founder mutations in BRCA1 and BRCA2 genes. Ann Oncol 2007; 18 Suppl 6:vi93.
  5. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet 2003; 72:1117.
  6. Begg CB, Haile RW, Borg A, et al. Variation of breast cancer risk among BRCA1/2 carriers. JAMA 2008; 299:194.
  7. Wang F, Fang Q, Ge Z, et al. Common BRCA1 and BRCA2 mutations in breast cancer families: a meta-analysis from systematic review. Mol Biol Rep 2012; 39:2109.
  8. Judkins T, Rosenthal E, Arnell C, et al. Clinical significance of large rearrangements in BRCA1 and BRCA2. Cancer 2012; 118:5210.
  9. McClain MR, Palomaki GE, Nathanson KL, Haddow JE. Adjusting the estimated proportion of breast cancer cases associated with BRCA1 and BRCA2 mutations: public health implications. Genet Med 2005; 7:28.
  10. Roa BB, Boyd AA, Volcik K, Richards CS. Ashkenazi Jewish population frequencies for common mutations in BRCA1 and BRCA2. Nat Genet 1996; 14:185.
  11. King MC, Marks JH, Mandell JB, New York Breast Cancer Study Group. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science 2003; 302:643.
  12. Gershoni-Baruch R, Dagan E, Fried G, et al. Significantly lower rates of BRCA1/BRCA2 founder mutations in Ashkenazi women with sporadic compared with familial early onset breast cancer. Eur J Cancer 2000; 36:983.
  13. Hodgson SV, Heap E, Cameron J, et al. Risk factors for detecting germline BRCA1 and BRCA2 founder mutations in Ashkenazi Jewish women with breast or ovarian cancer. J Med Genet 1999; 36:369.
  14. Kauff ND, Perez-Segura P, Robson ME, et al. Incidence of non-founder BRCA1 and BRCA2 mutations in high risk Ashkenazi breast and ovarian cancer families. J Med Genet 2002; 39:611.
  15. Phelan CM, Kwan E, Jack E, et al. A low frequency of non-founder BRCA1 mutations in Ashkenazi Jewish breast-ovarian cancer families. Hum Mutat 2002; 20:352.
  16. Frank TS, Deffenbaugh AM, Reid JE, et al. Clinical characteristics of individuals with germline mutations in BRCA1 and BRCA2: analysis of 10,000 individuals. J Clin Oncol 2002; 20:1480.
  17. Rosenthal E, Moyes K, Arnell C, et al. Incidence of BRCA1 and BRCA2 non-founder mutations in patients of Ashkenazi Jewish ancestry. Breast Cancer Res Treat 2015; 149:223.
  18. Thorlacius S, Sigurdsson S, Bjarnadottir H, et al. Study of a single BRCA2 mutation with high carrier frequency in a small population. Am J Hum Genet 1997; 60:1079.
  19. Rafnar T, Benediktsdottir KR, Eldon BJ, et al. BRCA2, but not BRCA1, mutations account for familial ovarian cancer in Iceland: a population-based study. Eur J Cancer 2004; 40:2788.
  20. Bergthorsson JT, Jonasdottir A, Johannesdottir G, et al. Identification of a novel splice-site mutation of the BRCA1 gene in two breast cancer families: screening reveals low frequency in Icelandic breast cancer patients. Hum Mutat 1998; Suppl 1:S195.
  21. Ghadirian P, Robidoux A, Zhang P, et al. The contribution of founder mutations to early-onset breast cancer in French-Canadian women. Clin Genet 2009; 76:421.
  22. Ghadirian P, Robidoux A, Nassif E, et al. Screening for BRCA1 and BRCA2 mutations among French-Canadian breast cancer cases attending an outpatient clinic in Montreal. Clin Genet 2014; 85:31.
  23. Weitzel JN, Clague J, Martir-Negron A, et al. Prevalence and type of BRCA mutations in Hispanics undergoing genetic cancer risk assessment in the southwestern United States: a report from the Clinical Cancer Genetics Community Research Network. J Clin Oncol 2013; 31:210.
  24. Malone KE, Daling JR, Doody DR, et al. Prevalence and predictors of BRCA1 and BRCA2 mutations in a population-based study of breast cancer in white and black American women ages 35 to 64 years. Cancer Res 2006; 66:8297.
  25. Fackenthal JD, Zhang J, Zhang B, et al. High prevalence of BRCA1 and BRCA2 mutations in unselected Nigerian breast cancer patients. Int J Cancer 2012; 131:1114.
  26. Gao Q, Tomlinson G, Das S, et al. Prevalence of BRCA1 and BRCA2 mutations among clinic-based African American families with breast cancer. Hum Genet 2000; 107:186.
  27. Gao Q, Adebamowo CA, Fackenthal J, et al. Protein truncating BRCA1 and BRCA2 mutations in African women with pre-menopausal breast cancer. Hum Genet 2000; 107:192.
  28. Nanda R, Schumm LP, Cummings S, et al. Genetic testing in an ethnically diverse cohort of high-risk women: a comparative analysis of BRCA1 and BRCA2 mutations in American families of European and African ancestry. JAMA 2005; 294:1925.
  29. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 2007; 25:1329.
  30. Chen J, Bae E, Zhang L, et al. Penetrance of Breast and Ovarian Cancer in Women Who Carry a BRCA1/2 Mutation and Do Not Use Risk-Reducing Salpingo-Oophorectomy: An Updated Meta-Analysis. JNCI Cancer Spectr 2020; 4:pkaa029.
  31. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 2017; 317:2402.
  32. Mavaddat N, Peock S, Frost D, et al. Cancer risks for BRCA1 and BRCA2 mutation carriers: results from prospective analysis of EMBRACE. J Natl Cancer Inst 2013; 105:812.
  33. van der Kolk DM, de Bock GH, Leegte BK, et al. Penetrance of breast cancer, ovarian cancer and contralateral breast cancer in BRCA1 and BRCA2 families: high cancer incidence at older age. Breast Cancer Res Treat 2010; 124:643.
  34. 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.
  35. Meijers-Heijboer EJ, Verhoog LC, Brekelmans CT, et al. Presymptomatic DNA testing and prophylactic surgery in families with a BRCA1 or BRCA2 mutation. Lancet 2000; 355:2015.
  36. Copson ER, Maishman TC, Tapper WJ, et al. Germline BRCA mutation and outcome in young-onset breast cancer (POSH): a prospective cohort study. Lancet Oncol 2018; 19:169.
  37. Tun NM, Villani G, Ong K, et al. Risk of having BRCA1 mutation in high-risk women with triple-negative breast cancer: a meta-analysis. Clin Genet 2014; 85:43.
  38. Hwang ES, McLennan JL, Moore DH, et al. Ductal carcinoma in situ in BRCA mutation carriers. J Clin Oncol 2007; 25:642.
  39. Smith KL, Adank M, Kauff N, et al. BRCA mutations in women with ductal carcinoma in situ. Clin Cancer Res 2007; 13:4306.
  40. Bayraktar S, Elsayegh N, Gutierrez Barrera AM, et al. Predictive factors for BRCA1/BRCA2 mutations in women with ductal carcinoma in situ. Cancer 2012; 118:1515.
  41. Hall MJ, Reid JE, Wenstrup RJ. Prevalence of BRCA1 and BRCA2 mutations in women with breast carcinoma In Situ and referred for genetic testing. Cancer Prev Res (Phila) 2010; 3:1579.
  42. Tai YC, Domchek S, Parmigiani G, Chen S. Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 2007; 99:1811.
  43. Evans DG, Susnerwala I, Dawson J, et al. Risk of breast cancer in male BRCA2 carriers. J Med Genet 2010; 47:710.
  44. Li S, Silvestri V, Leslie G, et al. Cancer Risks Associated With BRCA1 and BRCA2 Pathogenic Variants. J Clin Oncol 2022; 40:1529.
  45. DevCan version 6.7.9, April 2021, National Cancer Institute. Available at: https://surveillance.cancer.gov/devcan/ (Accessed on April 06, 2022).
  46. Brose MS, Rebbeck TR, Calzone KA, et al. Cancer risk estimates for BRCA1 mutation carriers identified in a risk evaluation program. J Natl Cancer Inst 2002; 94:1365.
  47. Aziz S, Kuperstein G, Rosen B, et al. A genetic epidemiological study of carcinoma of the fallopian tube. Gynecol Oncol 2001; 80:341.
  48. Levine DA, Argenta PA, Yee CJ, et al. Fallopian tube and primary peritoneal carcinomas associated with BRCA mutations. J Clin Oncol 2003; 21:4222.
  49. Stewart SL, Wike JM, Foster SL, Michaud F. The incidence of primary fallopian tube cancer in the United States. Gynecol Oncol 2007; 107:392.
  50. Lee Y, Miron A, Drapkin R, et al. A candidate precursor to serous carcinoma that originates in the distal fallopian tube. J Pathol 2007; 211:26.
  51. Reade CJ, McVey RM, Tone AA, et al. The fallopian tube as the origin of high grade serous ovarian cancer: review of a paradigm shift. J Obstet Gynaecol Can 2014; 36:133.
  52. Vicus D, Finch A, Cass I, et al. Prevalence of BRCA1 and BRCA2 germ line mutations among women with carcinoma of the fallopian tube. Gynecol Oncol 2010; 118:299.
  53. Thompson D, Easton DF, Breast Cancer Linkage Consortium. Cancer Incidence in BRCA1 mutation carriers. J Natl Cancer Inst 2002; 94:1358.
  54. Moslehi R, Chu W, Karlan B, et al. BRCA1 and BRCA2 mutation analysis of 208 Ashkenazi Jewish women with ovarian cancer. Am J Hum Genet 2000; 66:1259.
  55. Segev Y, Iqbal J, Lubinski J, et al. The incidence of endometrial cancer in women with BRCA1 and BRCA2 mutations: an international prospective cohort study. Gynecol Oncol 2013; 130:127.
  56. Shu CA, Pike MC, Jotwani AR, et al. Uterine Cancer After Risk-Reducing Salpingo-oophorectomy Without Hysterectomy in Women With BRCA Mutations. JAMA Oncol 2016; 2:1434.
  57. Kitson SJ, Bafligil C, Ryan NAJ, et al. BRCA1 and BRCA2 pathogenic variant carriers and endometrial cancer risk: A cohort study. Eur J Cancer 2020; 136:169.
  58. de Jonge MM, de Kroon CD, Jenner DJ, et al. Endometrial Cancer Risk in Women With Germline BRCA1 or BRCA2 Mutations: Multicenter Cohort Study. J Natl Cancer Inst 2021; 113:1203.
  59. Pennington KP, Walsh T, Lee M, et al. BRCA1, TP53, and CHEK2 germline mutations in uterine serous carcinoma. Cancer 2013; 119:332.
  60. Breast Cancer Linkage Consortium. Cancer risks in BRCA2 mutation carriers. J Natl Cancer Inst 1999; 91:1310.
  61. Ferrone CR, Levine DA, Tang LH, et al. BRCA germline mutations in Jewish patients with pancreatic adenocarcinoma. J Clin Oncol 2009; 27:433.
  62. Iqbal J, Ragone A, Lubinski J, et al. The incidence of pancreatic cancer in BRCA1 and BRCA2 mutation carriers. Br J Cancer 2012; 107:2005.
  63. Kim DH, Crawford B, Ziegler J, Beattie MS. Prevalence and characteristics of pancreatic cancer in families with BRCA1 and BRCA2 mutations. Fam Cancer 2009; 8:153.
  64. Golan T, Kanji ZS, Epelbaum R, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br J Cancer 2014; 111:1132.
  65. Kote-Jarai Z, Leongamornlert D, Saunders E, et al. BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients. Br J Cancer 2011; 105:1230.
  66. Leongamornlert D, Mahmud N, Tymrakiewicz M, et al. Germline BRCA1 mutations increase prostate cancer risk. Br J Cancer 2012; 106:1697.
  67. Lecarpentier J, Silvestri V, Kuchenbaecker KB, et al. Prediction of Breast and Prostate Cancer Risks in Male BRCA1 and BRCA2 Mutation Carriers Using Polygenic Risk Scores. J Clin Oncol 2017; 35:2240.
  68. Mitra AV, Bancroft EK, Barbachano Y, et al. Targeted prostate cancer screening in men with mutations in BRCA1 and BRCA2 detects aggressive prostate cancer: preliminary analysis of the results of the IMPACT study. BJU Int 2011; 107:28.
  69. Castro E, Goh C, Olmos D, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol 2013; 31:1748.
  70. Mitra A, Fisher C, Foster CS, et al. Prostate cancer in male BRCA1 and BRCA2 mutation carriers has a more aggressive phenotype. Br J Cancer 2008; 98:502.
  71. Thorne H, Willems AJ, Niedermayr E, et al. Decreased prostate cancer-specific survival of men with BRCA2 mutations from multiple breast cancer families. Cancer Prev Res (Phila) 2011; 4:1002.
  72. Mersch J, Jackson MA, Park M, et al. Cancers associated with BRCA1 and BRCA2 mutations other than breast and ovarian. Cancer 2015; 121:269.
  73. Gumaste PV, Penn LA, Cymerman RM, et al. Skin cancer risk in BRCA1/2 mutation carriers. Br J Dermatol 2015; 172:1498.
  74. Moran A, O'Hara C, Khan S, et al. Risk of cancer other than breast or ovarian in individuals with BRCA1 and BRCA2 mutations. Fam Cancer 2012; 11:235.
  75. Cruz C, Teule A, Caminal JM, et al. Uveal melanoma and BRCA1/BRCA2 genes: a relationship that needs further investigation. J Clin Oncol 2011; 29:e827.
  76. Momozawa Y, Sasai R, Usui Y, et al. Expansion of Cancer Risk Profile for BRCA1 and BRCA2 Pathogenic Variants. JAMA Oncol 2022; 8:871.
  77. Schrag D, Kuntz KM, Garber JE, Weeks JC. Decision analysis--effects of prophylactic mastectomy and oophorectomy on life expectancy among women with BRCA1 or BRCA2 mutations. N Engl J Med 1997; 336:1465.
  78. Schrag D, Kuntz KM, Garber JE, Weeks JC. Life expectancy gains from cancer prevention strategies for women with breast cancer and BRCA1 or BRCA2 mutations. JAMA 2000; 283:617.
  79. Grann VR, Jacobson JS, Thomason D, et al. Effect of prevention strategies on survival and quality-adjusted survival of women with BRCA1/2 mutations: an updated decision analysis. J Clin Oncol 2002; 20:2520.
  80. van Roosmalen MS, Stalmeier PF, Verhoef LC, et al. Randomized trial of a shared decision-making intervention consisting of trade-offs and individualized treatment information for BRCA1/2 mutation carriers. J Clin Oncol 2004; 22:3293.
  81. Kurian AW, Sigal BM, Plevritis SK. Survival analysis of cancer risk reduction strategies for BRCA1/2 mutation carriers. J Clin Oncol 2010; 28:222.
  82. Friebel TM, Domchek SM, Rebbeck TR. Modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: systematic review and meta-analysis. J Natl Cancer Inst 2014; 106:dju091.
  83. Genetic/familial high-risk assessment: colorectal 2.2022. NCCN clinical practice guidelines in oncology. Available at: https://www.nccn.org/login?ReturnURL=https://www.nccn.org/professionals/physician_gls/pdf/genetics_colon.pdf (Accessed on March 21, 2023).
  84. Meijers-Heijboer H, van Geel B, van Putten WL, et al. Breast cancer after prophylactic bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2001; 345:159.
  85. Rebbeck TR, Friebel T, Lynch HT, et al. Bilateral prophylactic mastectomy reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE Study Group. J Clin Oncol 2004; 22:1055.
  86. Hartmann LC, Schaid DJ, Woods JE, et al. Efficacy of bilateral prophylactic mastectomy in women with a family history of breast cancer. N Engl J Med 1999; 340:77.
  87. Hartmann LC, Sellers TA, Schaid DJ, et al. Efficacy of bilateral prophylactic mastectomy in BRCA1 and BRCA2 gene mutation carriers. J Natl Cancer Inst 2001; 93:1633.
  88. Heemskerk-Gerritsen BA, Brekelmans CT, Menke-Pluymers MB, et al. Prophylactic mastectomy in BRCA1/2 mutation carriers and women at risk of hereditary breast cancer: long-term experiences at the Rotterdam Family Cancer Clinic. Ann Surg Oncol 2007; 14:3335.
  89. Geiger AM, Yu O, Herrinton LJ, et al. A population-based study of bilateral prophylactic mastectomy efficacy in women at elevated risk for breast cancer in community practices. Arch Intern Med 2005; 165:516.
  90. 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.
  91. Ingham SL, Sperrin M, Baildam A, et al. Risk-reducing surgery increases survival in BRCA1/2 mutation carriers unaffected at time of family referral. Breast Cancer Res Treat 2013; 142:611.
  92. Ludwig KK, Neuner J, Butler A, et al. Risk reduction and survival benefit of prophylactic surgery in BRCA mutation carriers, a systematic review. Am J Surg 2016; 212:660.
  93. Metcalfe K, Huzarski T, Gronwald J, et al. Risk-reducing mastectomy and breast cancer mortality in women with a BRCA1 or BRCA2 pathogenic variant: an international analysis. Br J Cancer 2024; 130:269.
  94. Jakub JW, Peled AW, Gray RJ, et al. Oncologic Safety of Prophylactic Nipple-Sparing Mastectomy in a Population With BRCA Mutations: A Multi-institutional Study. JAMA Surg 2018; 153:123.
  95. Garstka M, Henriquez A, Kelly BN, et al. How Protective are Nipple-Sparing Prophylactic Mastectomies in BRCA1 and BRCA2 Mutation Carriers? Ann Surg Oncol 2021; 28:5657.
  96. Scheuer L, Kauff N, Robson M, et al. Outcome of preventive surgery and screening for breast and ovarian cancer in BRCA mutation carriers. J Clin Oncol 2002; 20:1260.
  97. Rebbeck TR, Lynch HT, Neuhausen SL, et al. Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 2002; 346:1616.
  98. Kauff ND, Satagopan JM, Robson ME, et al. Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 2002; 346:1609.
  99. Haber D. Prophylactic oophorectomy to reduce the risk of ovarian and breast cancer in carriers of BRCA mutations. N Engl J Med 2002; 346:1660.
  100. Marchetti C, De Felice F, Palaia I, et al. Risk-reducing salpingo-oophorectomy: a meta-analysis on impact on ovarian cancer risk and all cause mortality in BRCA 1 and BRCA 2 mutation carriers. BMC Womens Health 2014; 14:150.
  101. Struewing JP, Watson P, Easton DF, et al. Prophylactic oophorectomy in inherited breast/ovarian cancer families. J Natl Cancer Inst Monogr 1995; :33.
  102. Menczer J, Chetrit A, Barda G, et al. Frequency of BRCA mutations in primary peritoneal carcinoma in Israeli Jewish women. Gynecol Oncol 2003; 88:58.
  103. Karlan BY, Baldwin RL, Lopez-Luevanos E, et al. Peritoneal serous papillary carcinoma, a phenotypic variant of familial ovarian cancer: implications for ovarian cancer screening. Am J Obstet Gynecol 1999; 180:917.
  104. Steenbeek MP, van Bommel MHD, Bulten J, et al. Risk of Peritoneal Carcinomatosis After Risk-Reducing Salpingo-Oophorectomy: A Systematic Review and Individual Patient Data Meta-Analysis. J Clin Oncol 2022; 40:1879.
  105. Eisen A, Rebbeck TR, Wood WC, Weber BL. Prophylactic surgery in women with a hereditary predisposition to breast and ovarian cancer. J Clin Oncol 2000; 18:1980.
  106. Zhou J, Iwasa Y, Konishi I, et al. Papillary serous carcinoma of the peritoneum in women. A clinicopathologic and immunohistochemical study. Cancer 1995; 76:429.
  107. Dowdy SC, Stefanek M, Hartmann LC. Surgical risk reduction: prophylactic salpingo-oophorectomy and prophylactic mastectomy. Am J Obstet Gynecol 2004; 191:1113.
  108. Finch A, Beiner M, Lubinski J, et al. Salpingo-oophorectomy and the risk of ovarian, fallopian tube, and peritoneal cancers in women with a BRCA1 or BRCA2 Mutation. JAMA 2006; 296:185.
  109. Hilton JL, Geisler JP, Rathe JA, et al. Inactivation of BRCA1 and BRCA2 in ovarian cancer. J Natl Cancer Inst 2002; 94:1396.
  110. Casey MJ, Synder C, Bewtra C, et al. Intra-abdominal carcinomatosis after prophylactic oophorectomy in women of hereditary breast ovarian cancer syndrome kindreds associated with BRCA1 and BRCA2 mutations. Gynecol Oncol 2005; 97:457.
  111. Choi YH, Terry MB, Daly MB, et al. Association of Risk-Reducing Salpingo-Oophorectomy With Breast Cancer Risk in Women With BRCA1 and BRCA2 Pathogenic Variants. JAMA Oncol 2021; 7:585.
  112. Kotsopoulos J, Huzarski T, Gronwald J, et al. Bilateral Oophorectomy and Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst 2017; 109.
  113. Biron-Shental T, Drucker L, Altaras M, et al. High incidence of BRCA1-2 germline mutations, previous breast cancer and familial cancer history in Jewish patients with uterine serous papillary carcinoma. Eur J Surg Oncol 2006; 32:1097.
  114. SGO clinical practice statement: Salpingectomy for ovarian cancer prevention. Available at: www.sgo.org/clinical-practice/guidelines/sgo-clinical-practice-statement-salpingectomy-for-ovarian-cancer-prevention/ (Accessed on December 11, 2019).
  115. Steenbeek MP, Harmsen MG, Hoogerbrugge N, et al. Association of Salpingectomy With Delayed Oophorectomy Versus Salpingo-oophorectomy With Quality of Life in BRCA1/2 Pathogenic Variant Carriers: A Nonrandomized Controlled Trial. JAMA Oncol 2021; 7:1203.
  116. Kurman RJ, Shih IeM. The origin and pathogenesis of epithelial ovarian cancer: a proposed unifying theory. Am J Surg Pathol 2010; 34:433.
  117. Greene MH, Mai PL, Schwartz PE. Does bilateral salpingectomy with ovarian retention warrant consideration as a temporary bridge to risk-reducing bilateral oophorectomy in BRCA1/2 mutation carriers? Am J Obstet Gynecol 2011; 204:19.e1.
  118. Walker JL, Powell CB, Chen LM, et al. Society of Gynecologic Oncology recommendations for the prevention of ovarian cancer. Cancer 2015; 121:2108.
  119. Sinno AK, Pinkerton J, Febbraro T, et al. Hormone therapy (HT) in women with gynecologic cancers and in women at high risk for developing a gynecologic cancer: A Society of Gynecologic Oncology (SGO) clinical practice statement: This practice statement has been endorsed by The North American Menopause Society. Gynecol Oncol 2020; 157:303.
  120. Kotsopoulos J, Gronwald J, Karlan BY, et al. Hormone Replacement Therapy After Oophorectomy and Breast Cancer Risk Among BRCA1 Mutation Carriers. JAMA Oncol 2018; 4:1059.
  121. Rebbeck TR, Friebel T, Wagner T, et al. Effect of short-term hormone replacement therapy on breast cancer risk reduction after bilateral prophylactic oophorectomy in BRCA1 and BRCA2 mutation carriers: the PROSE Study Group. J Clin Oncol 2005; 23:7804.
  122. Domchek SM, Friebel T, Neuhausen SL, et al. Is hormone replacement therapy (HRT) following risk-reducing salpingectomy (RRSO) in BRCA1- (B1)- and BRCA-2 (B2)-mutation carriers associated with an increased risk of breast cancer? J Clin Oncol 2011; 29S: ASCO #1501.
  123. Michaelson-Cohen R, Gabizon-Peretz S, Armon S, et al. Breast cancer risk and hormone replacement therapy among BRCA carriers after risk-reducing salpingo-oophorectomy. Eur J Cancer 2021; 148:95.
  124. Holmberg L, Iversen OE, Rudenstam CM, et al. Increased risk of recurrence after hormone replacement therapy in breast cancer survivors. J Natl Cancer Inst 2008; 100:475.
  125. Saslow D, Boetes C, Burke W, et al. American Cancer Society guidelines for breast screening with MRI as an adjunct to mammography. CA Cancer J Clin 2007; 57:75.
  126. Le-Petross HT, Whitman GJ, Atchley DP, et al. Effectiveness of alternating mammography and magnetic resonance imaging for screening women with deleterious BRCA mutations at high risk of breast cancer. Cancer 2011; 117:3900.
  127. Brekelmans CT, Seynaeve C, Bartels CC, et al. Effectiveness of breast cancer surveillance in BRCA1/2 gene mutation carriers and women with high familial risk. J Clin Oncol 2001; 19:924.
  128. Tilanus-Linthorst MM, Obdeijn IM, Bartels KC, et al. First experiences in screening women at high risk for breast cancer with MR imaging. Breast Cancer Res Treat 2000; 63:53.
  129. Ziv E, Shepherd J, Smith-Bindman R, Kerlikowske K. Mammographic breast density and family history of breast cancer. J Natl Cancer Inst 2003; 95:556.
  130. Komenaka IK, Ditkoff BA, Joseph KA, et al. The development of interval breast malignancies in patients with BRCA mutations. Cancer 2004; 100:2079.
  131. Kriege M, Brekelmans CT, Boetes C, et al. Efficacy of MRI and mammography for breast-cancer screening in women with a familial or genetic predisposition. N Engl J Med 2004; 351:427.
  132. Warner E, Hill K, Causer P, et al. Prospective study of breast cancer incidence in women with a BRCA1 or BRCA2 mutation under surveillance with and without magnetic resonance imaging. J Clin Oncol 2011; 29:1664.
  133. Plevritis SK, Kurian AW, Sigal BM, et al. Cost-effectiveness of screening BRCA1/2 mutation carriers with breast magnetic resonance imaging. JAMA 2006; 295:2374.
  134. Monticciolo DL, Newell MS, Moy L, et al. Breast Cancer Screening for Women at Higher-Than-Average Risk: Updated Recommendations From the ACR. J Am Coll Radiol 2023; 20:902.
  135. Warner E, Messersmith H, Causer P, et al. Systematic review: using magnetic resonance imaging to screen women at high risk for breast cancer. Ann Intern Med 2008; 148:671.
  136. Pijpe A, Andrieu N, Easton DF, et al. Exposure to diagnostic radiation and risk of breast cancer among carriers of BRCA1/2 mutations: retrospective cohort study (GENE-RAD-RISK). BMJ 2012; 345:e5660.
  137. Narod SA, Lubinski J, Ghadirian P, et al. Screening mammography and risk of breast cancer in BRCA1 and BRCA2 mutation carriers: a case-control study. Lancet Oncol 2006; 7:402.
  138. Andrieu N, Easton DF, Chang-Claude J, et al. Effect of chest X-rays on the risk of breast cancer among BRCA1/2 mutation carriers in the international BRCA1/2 carrier cohort study: a report from the EMBRACE, GENEPSO, GEO-HEBON, and IBCCS Collaborators' Group. J Clin Oncol 2006; 24:3361.
  139. Gronwald J, Pijpe A, Byrski T, et al. Early radiation exposures and BRCA1-associated breast cancer in young women from Poland. Breast Cancer Res Treat 2008; 112:581.
  140. Lehman CD, Isaacs C, Schnall MD, et al. Cancer yield of mammography, MR, and US in high-risk women: prospective multi-institution breast cancer screening study. Radiology 2007; 244:381.
  141. Sardanelli F, Podo F, Santoro F, et al. Multicenter surveillance of women at high genetic breast cancer risk using mammography, ultrasonography, and contrast-enhanced magnetic resonance imaging (the high breast cancer risk italian 1 study): final results. Invest Radiol 2011; 46:94.
  142. Riedl CC, Luft N, Bernhart C, et al. Triple-modality screening trial for familial breast cancer underlines the importance of magnetic resonance imaging and questions the role of mammography and ultrasound regardless of patient mutation status, age, and breast density. J Clin Oncol 2015; 33:1128.
  143. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Genetic/Familial High-risk Assessment: Breast and Ovarian. Version 2.2019. Available at: www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf (Accessed on September 10, 2018).
  144. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. National Comprehensive Cancer Network. Available at: https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf (Accessed on January 11, 2023).
  145. King MC, Wieand S, Hale K, et al. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA 2001; 286:2251.
  146. Eisen A, Weber BL. Prophylactic mastectomy for women with BRCA1 and BRCA2 mutations--facts and controversy. N Engl J Med 2001; 345:207.
  147. Vogel VG, Costantino JP, Wickerham DL, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 2006; 295:2727.
  148. Goss PE, Ingle JN, Alés-Martínez JE, et al. Exemestane for breast-cancer prevention in postmenopausal women. N Engl J Med 2011; 364:2381.
  149. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial. Lancet 2014; 383:1041.
  150. Iodice S, Barile M, Rotmensz N, et al. Oral contraceptive use and breast or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Cancer 2010; 46:2275.
  151. McLaughlin JR, Risch HA, Lubinski J, et al. Reproductive risk factors for ovarian cancer in carriers of BRCA1 or BRCA2 mutations: a case-control study. Lancet Oncol 2007; 8:26.
  152. Jordan SJ, Green AC, Whiteman DC, et al. Serous ovarian, fallopian tube and primary peritoneal cancers: a comparative epidemiological analysis. Int J Cancer 2008; 122:1598.
  153. Wang ET, Pisarska MD, Bresee C, et al. BRCA1 germline mutations may be associated with reduced ovarian reserve. Fertil Steril 2014; 102:1723.
  154. Son KA, Lee DY, Choi D. Association of BRCA Mutations and Anti-müllerian Hormone Level in Young Breast Cancer Patients. Front Endocrinol (Lausanne) 2019; 10:235.
  155. Johnson L, Sammel MD, Domchek S, et al. Antimüllerian hormone levels are lower in BRCA2 mutation carriers. Fertil Steril 2017; 107:1256.
  156. Gunnala V, Fields J, Irani M, et al. BRCA carriers have similar reproductive potential at baseline to noncarriers: comparisons in cancer and cancer-free cohorts undergoing fertility preservation. Fertil Steril 2019; 111:363.
  157. Turan V, Lambertini M, Lee DY, et al. Association of Germline BRCA Pathogenic Variants With Diminished Ovarian Reserve: A Meta-Analysis of Individual Patient-Level Data. J Clin Oncol 2021; 39:2016.
  158. Giri VN, Knudsen KE, Kelly WK, et al. Role of Genetic Testing for Inherited Prostate Cancer Risk: Philadelphia Prostate Cancer Consensus Conference 2017. J Clin Oncol 2018; 36:414.
  159. Silvestri V, Leslie G, Barnes DR, et al. Characterization of the Cancer Spectrum in Men With Germline BRCA1 and BRCA2 Pathogenic Variants: Results From the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). JAMA Oncol 2020; 6:1218.
  160. Sagi M, Weinberg N, Eilat A, et al. Preimplantation genetic diagnosis for BRCA1/2--a novel clinical experience. Prenat Diagn 2009; 29:508.
  161. Offit K, Levran O, Mullaney B, et al. Shared genetic susceptibility to breast cancer, brain tumors, and Fanconi anemia. J Natl Cancer Inst 2003; 95:1548.
  162. Sawyer SL, Tian L, Kähkönen M, et al. Biallelic mutations in BRCA1 cause a new Fanconi anemia subtype. Cancer Discov 2015; 5:135.
  163. Rahman N, Scott RH. Cancer genes associated with phenotypes in monoallelic and biallelic mutation carriers: new lessons from old players. Hum Mol Genet 2007; 16 Spec No 1:R60.
Topic 758 Version 71.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟