Skip to main content

Genetics of Breast and Gynecologic Cancers (PDQ®)–Health Professional Version

Executive Summary

This executive summary reviews the topics covered in this PDQ summary on the genetics of breast and gynecologic cancers, with hyperlinks to detailed sections below that describe the evidence on each topic.

  • Associated Genes and Syndromes

    Breast and ovarian cancer are present in several autosomal dominant cancer syndromes, although they are most strongly associated with highly penetrant germline pathogenic variants in BRCA1 and BRCA2. Other genes, such as PALB2, TP53 (associated with Li-Fraumeni syndrome), PTEN (associated with Cowden syndrome), CDH1 (associated with diffuse gastric and lobular breast cancer syndrome), and STK11 (associated with Peutz-Jeghers syndrome), confer a risk to either or both of these cancers with relatively high penetrance.

    Inherited endometrial cancer is most commonly associated with Lynch syndrome, a condition caused by inherited pathogenic variants in the highly penetrant mismatch repair genes MLH1, MSH2, MSH6, PMS2, and EPCAM. Colorectal cancer (and, to a lesser extent, ovarian cancer and stomach cancer) is also associated with Lynch syndrome.

    Additional genes, such as CHEK2, BRIP1, RAD51, and ATM, are associated with breast and/or gynecologic cancers with moderate penetrance. Genome-wide searches are showing promise in identifying common, low-penetrance susceptibility alleles for many complex diseases, including breast and gynecologic cancers, but the clinical utility of these findings remains uncertain.

  • Clinical Management

    Breast cancer screening strategies, including breast magnetic resonance imaging and mammography, are commonly performed in carriers of BRCA pathogenic variants and in individuals at increased risk of breast cancer. Initiation of screening is generally recommended at earlier ages and at more frequent intervals in individuals with an increased risk due to genetics and family history than in the general population. There is evidence to demonstrate that these strategies have utility in early detection of cancer. In contrast, there is currently no evidence to demonstrate that gynecologic cancer screening using cancer antigen 125 testing and transvaginal ultrasound leads to early detection of cancer.

    Risk-reducing surgeries, including risk-reducing mastectomy (RRM) and risk-reducing salpingo-oophorectomy (RRSO), have been shown to significantly reduce the risk of developing breast and/or ovarian cancer and improve overall survival in carriers of BRCA1 and BRCA2 pathogenic variants. Chemoprevention strategies, including the use of tamoxifen and oral contraceptives, have also been examined in this population. Tamoxifen use has been shown to reduce the risk of contralateral breast cancer among carriers of BRCA1 and BRCA2 pathogenic variants after treatment for breast cancer, but there are limited data in the primary cancer prevention setting to suggest that it reduces the risk of breast cancer among healthy female carriers of BRCA2 pathogenic variants. The use of oral contraceptives has been associated with a protective effect on the risk of developing ovarian cancer, including in carriers of BRCA1 and BRCA2 pathogenic variants, with no association of increased risk of breast cancer when using formulations developed after 1975.

  • Psychosocial and Behavioral Issues

    Psychosocial factors influence decisions about genetic testing for inherited cancer risk and risk-management strategies. Uptake of genetic testing varies widely across studies. Psychological factors that have been associated with testing uptake include cancer-specific distress and perceived risk of developing breast or ovarian cancer. Studies have shown low levels of distress after genetic testing for both carriers and noncarriers, particularly in the longer term. Uptake of RRM and RRSO also varies across studies and may be influenced by factors such as cancer history, age, family history, recommendations of the health care provider, and pretreatment genetic education and counseling. Patients' communication with their family members about an inherited risk of breast and gynecologic cancer is complex; gender, age, and the degree of relatedness are some elements that affect disclosure of this information. Research is ongoing to better understand and address psychosocial and behavioral issues in high-risk families.

Introduction

General Information

[Note: Many of the medical and scientific terms used in this summary are found in the NCI Dictionary of Genetics Terms. When a linked term is clicked, the definition will appear in a separate window.]

[Note: A concerted effort is being made within the genetics community to shift terminology used to describe genetic variation. The shift is to use the term “variant” rather than the term “mutation” to describe a genetic difference that exists between the person or group being studied and the reference sequence, particularly for differences that exist in the germline. Variants can then be further classified as benign (harmless), likely benign, of uncertain significance, likely pathogenic, or pathogenic (disease causing). Throughout this summary, we will use the term pathogenic variant to describe a disease-causing mutation. Refer to the Cancer Genetics Overview summary for more information about variant classification.]

[Note: Many of the genes and conditions described in this summary are found in the Online Mendelian Inheritance in Man (OMIM) catalog. Refer to OMIM for more information.]

Among women in the United States, breast cancer is the most commonly diagnosed cancer after nonmelanoma skin cancer, and it is the second leading cause of cancer deaths after lung cancer. In 2022, an estimated 290,560 new cases of breast cancer (including 2,710 cases in men) will be diagnosed, and 43,780 deaths (including 530 deaths in men) will occur.[1] The incidence of breast cancer, particularly for estrogen receptor (ER)–positive cancers occurring after age 50 years, is declining and has declined at a faster rate since 2003; this may be temporally related to a decrease in hormone replacement therapy (HRT) after early reports from the Women’s Health Initiative (WHI).[2] An estimated 19,880 new cases of ovarian cancer are expected in the United States in 2022, with an estimated 12,810 deaths. Ovarian cancer is the fifth most deadly cancer in women.[1] An estimated 65,950 new cases of endometrial cancer are expected in the United States in 2022, with an estimated 12,550 deaths.[1] (Refer to the PDQ summaries on Breast Cancer Treatment [Adult]; Ovarian Epithelial, Fallopian Tube, and Primary Peritoneal Cancer Treatment; and Endometrial Cancer Treatment for more information about breast, ovarian, and endometrial cancer rates, diagnosis, and management.)

A possible genetic contribution to both breast and ovarian cancer risk is indicated by the increased incidence of these cancers among women with a family history (refer to the Risk Factors for Breast Cancer, Risk Factors for Ovarian Cancer, and Risk Factors for Endometrial Cancer sections below for more information), and by the observation of some families in which multiple family members are affected with breast and/or ovarian cancer, in a pattern compatible with an inheritance of autosomal dominant cancer susceptibility. Formal studies of families (linkage analysis) have subsequently proven the existence of autosomal dominant predispositions to breast and ovarian cancer and have led to the identification of several highly penetrant genes as the cause of inherited cancer risk in many families. (Refer to the PDQ summary Cancer Genetics Overview for more information about linkage analysis.) Pathogenic variants in these genes are rare in the general population and are estimated to account for no more than 5% to 10% of breast and ovarian cancer cases overall. It is likely that other genetic factors contribute to the etiology of some of these cancers.

Risk Factors for Breast Cancer

Refer to the PDQ summary on Breast Cancer Prevention for information about risk factors for breast cancer in the general population.

Age

The cumulative risk of breast cancer increases with age, with most breast cancers occurring after age 50 years.[3] Breast cancer (and ovarian cancer, to a lesser degree) tends to occur at an earlier age in women with a genetic susceptibility than it does in women with sporadic cases.

Family history including inherited cancer genes

In cross-sectional studies of adult populations, 5% to 10% of women have a mother or sister with breast cancer, and about twice as many have either a first-degree relative (FDR) or a second-degree relative with breast cancer.[4-7] The risk conferred by a family history of breast cancer has been assessed in case-control and cohort studies, using volunteer and population-based samples, with generally consistent results.[8] In a pooled analysis of 38 studies, the relative risk (RR) of breast cancer conferred by an FDR with breast cancer was 2.1 (95% confidence interval [CI], 2.0–2.2).[8] Risk increases with the number of affected relatives, age at diagnosis, the occurrence of bilateral or multiple ipsilateral breast cancers in a family member, and the number of affected male relatives.[5,6,8-10] A large population-based study from the Swedish Family Cancer Database confirmed the finding of a significantly increased risk of breast cancer in women who had a mother or a sister with breast cancer. The hazard ratio (HR) for women with a single breast cancer in the family was 1.8 (95% CI, 1.8–1.9) and was 2.7 (95% CI, 2.6–2.9) for women with a family history of multiple breast cancers. For women who had multiple breast cancers in the family, with one occurring before age 40 years, the HR was 3.8 (95% CI, 3.1–4.8). However, the study also found a significant increase in breast cancer risk if the relative was aged 60 years or older, suggesting that breast cancer at any age in the family carries some increase in risk.[10] Another study in women with unilateral versus contralateral breast cancer (CBC) evaluated breast cancer risk among family members.[11] Results indicated that among women with affected FDRs, CBC risk was 8.1% at 10 years. This risk was higher among relatives diagnosed before age 40 years or with CBC and approached the lower risk estimates among BRCA carriers. (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for information about cancer risk estimates in that population.) These risk estimates remained unchanged when the analysis was restricted to women who tested negative for a deleterious variant in BRCA1/BRCA2, ATM, CHEK2, and PALB2.

One of the largest studies of twins ever conducted, with 80,309 monozygotic twins and 123,382 dizygotic twins, reported a heritability estimate for breast cancer of 31% (95% CI, 11%–51%).[12] If a monozygotic twin had breast cancer, her twin sister had a 28.1% probability of developing breast cancer (95% CI, 23.9%–32.8%), and if a dizygotic twin had breast cancer, her twin sister had a 19.9% probability of developing breast cancer (95% CI, 17%–23.2%). These estimates suggest a 10% higher risk of breast cancer for monozygotic twins than for dizygotic twins. However, a high rate of discordance even between monozygotic twins suggests that environmental factors also have a role in modifying breast cancer risk.

(Refer to the Penetrance of BRCA pathogenic variants section of this summary for a discussion of familial risk in women from families with BRCA1/BRCA2 pathogenic variants who themselves test negative for the family pathogenic variant.)

Reproductive history

In general, breast cancer risk increases with early menarche and late menopause and is reduced by early first full-term pregnancy. There may be an increased risk of breast cancer in carriers of BRCA1 and BRCA2 pathogenic variants with pregnancy at a younger age (before age 30 y), with a more significant effect seen for carriers of BRCA1 pathogenic variants.[13-15] Likewise, breastfeeding can reduce breast cancer risk in carriers of BRCA1 (but not BRCA2) pathogenic variants.[16] Regarding the effect of pregnancy on breast cancer outcomes, neither diagnosis of breast cancer during pregnancy nor pregnancy after breast cancer seems to be associated with adverse survival outcomes in women who carry a BRCA1 or BRCA2 pathogenic variant.[17] Parity appears to be protective for carriers of BRCA1 and BRCA2 pathogenic variants, with an additional protective effect for live birth before age 40 years.[18]

Reproductive history can also affect the risk of ovarian cancer and endometrial cancer. (Refer to the Reproductive History sections in the Risk Factors for Ovarian Cancer and Risk Factors for Endometrial Cancer sections of this summary for more information.)

Oral contraceptives

Oral contraceptives (OCs) may produce a slight increase in breast cancer risk among long-term users, but this appears to be a short-term effect. In a meta-analysis of data from 54 studies, the risk of breast cancer associated with OC use did not vary in relationship to a family history of breast cancer.[19]

OCs are sometimes recommended for ovarian cancer prevention in carriers of BRCA1 and BRCA2 pathogenic variants. (Refer to the Oral Contraceptives section in the Risk Factors for Ovarian Cancer section of this summary for more information.) Although the data are not entirely consistent, a meta-analysis concluded that there was no significant increased risk of breast cancer with OC use in carriers of BRCA1/BRCA2 pathogenic variants.[20] However, use of OCs formulated before 1975 was associated with an increased risk of breast cancer (summary relative risk [SRR], 1.47; 95% CI, 1.06–2.04).[20] (Refer to the Reproductive factors section in the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information.)

Hormone replacement therapy

Data exist from both observational and randomized clinical trials regarding the association between postmenopausal HRT and breast cancer. A meta-analysis of data from 51 observational studies indicated a RR of breast cancer of 1.35 (95% CI, 1.21–1.49) for women who had used HRT for 5 or more years after menopause.[21] The WHI (NCT00000611), a randomized controlled trial of about 160,000 postmenopausal women, investigated the risks and benefits of HRT. The estrogen-plus-progestin arm of the study, in which more than 16,000 women were randomly assigned to receive combined HRT or placebo, was halted early because health risks exceeded benefits.[22,23] Adverse outcomes prompting closure included significant increase in both total (245 vs. 185 cases) and invasive (199 vs. 150 cases) breast cancers (RR, 1.24; 95% CI, 1.02–1.5, P < . 001) and increased risks of coronary heart disease, stroke, and pulmonary embolism. Similar findings were seen in the estrogen-progestin arm of the prospective observational Million Women’s Study in the United Kingdom.[24] The risk of breast cancer was not elevated, however, in women randomly assigned to estrogen-only versus placebo in the WHI study (RR, 0.77; 95% CI, 0.59–1.01). Eligibility for the estrogen-only arm of this study required hysterectomy, and 40% of these patients also had undergone oophorectomy, which potentially could have impacted breast cancer risk.[25]

The association between HRT and breast cancer risk among women with a family history of breast cancer has not been consistent; some studies suggest risk is particularly elevated among women with a family history, while others have not found evidence for an interaction between these factors.[26-30,21] The increased risk of breast cancer associated with HRT use in the large meta-analysis did not differ significantly between subjects with and without a family history.[30] The WHI study has not reported analyses stratified on breast cancer family history, and subjects have not been systematically tested for BRCA1/BRCA2 pathogenic variants.[23] Short-term use of hormones for treatment of menopausal symptoms appears to confer little or no breast cancer risk.[21,31] The effect of HRT on breast cancer risk among carriers of BRCA1 or BRCA2 pathogenic variants has been studied in the context of bilateral risk-reducing oophorectomy, in which short-term replacement does not appear to reduce the protective effect of oophorectomy on breast cancer risk.[32] (Refer to the Hormone replacement therapy in carriers of BRCA1/BRCA2 pathogenic variants section of this summary for more information.)

Hormone use can also affect the risk of developing endometrial cancer. (Refer to the Hormones section in the Risk Factors for Endometrial Cancer section of this summary for more information.)

Radiation exposure

Observations in survivors of the atomic bombings of Hiroshima and Nagasaki and in women who have received therapeutic radiation treatments to the chest and upper body document increased breast cancer risk as a result of radiation exposure. The significance of this risk factor in women with a genetic susceptibility to breast cancer is unclear.

Preliminary data suggest that increased sensitivity to radiation could be a cause of cancer susceptibility in carriers of BRCA1 or BRCA2 pathogenic variants,[33-36] and in association with germline ATM and TP53 variants.[37,38]

The possibility that genetic susceptibility to breast cancer occurs via a mechanism of radiation sensitivity raises questions about radiation exposure. It is possible that diagnostic radiation exposure, including mammography, poses more risk in genetically susceptible women than in women of average risk. Therapeutic radiation could also pose a carcinogenic risk. A cohort study of carriers of BRCA1 and BRCA2 pathogenic variants treated with breast-conserving therapy, however, showed no evidence of increased radiation sensitivity or sequelae in the breast, lung, or bone marrow of carriers.[39] This finding was confirmed in a retrospective cohort study of 691 patients with BRCA1/BRCA2-associated breast cancer who were followed up for a median of 8.6 years. No association between receiving adjuvant radiation therapy and increased risk of CBC was observed in the entire cohort, including the subset of patients younger than 40 years at primary breast cancer diagnosis.[40] Conversely, radiation sensitivity could make tumors in women with genetic susceptibility to breast cancer more responsive to radiation treatment. Studies examining the impact of radiation exposure, including, but not limited to, mammography, in carriers of BRCA1 and BRCA2 pathogenic variants have had conflicting results.[41-46] A large European study showed a dose-response relationship of increased risk with total radiation exposure, but this was primarily driven by nonmammographic radiation exposure before age 20 years.[45] Subsequently, no significant association was observed between prior mammography exposure and breast cancer risk in a prospective study of 1,844 BRCA1 carriers and 502 BRCA2 carriers without a breast cancer diagnosis at time of study entry; average follow-up time was 5.3 years.[46] (Refer to the Mammography section in the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information about radiation.)

Alcohol intake

The risk of breast cancer increases by approximately 10% for each 10 g of daily alcohol intake (approximately one drink or less) in the general population.[47,48] Prior studies of carriers of BRCA1/BRCA2 pathogenic variants have found no increased risk associated with alcohol consumption.[49-51]

Physical activity and anthropometry

Increased physical activity has been associated with reduced breast cancer risk in most epidemiological studies. The reduction of risk has also been seen in studies of women with BRCA1 or BRCA2 pathogenic variants. For example, one study reported a 38% reduction in premenopausal breast cancer risk from moderate physical activity (odds ratio [OR] for the top quartile of physical activity compared with the lowest level, 0.62; 95% CI, 0.40–0.96).[52] The reduction of breast cancer risk has also been seen across the spectrum of absolute breast cancer risk, including in women who are at higher risk for breast cancer on the basis of their breast cancer family history but who do not have known BRCA1 or BRCA2 pathogenic variants.[53]

Benign breast disease and mammographic density

Benign breast disease (BBD) is a risk factor for breast cancer, independent of the effects of other major risk factors for breast cancer (age, age at menarche, age at first live birth, and family history of breast cancer).[54] There may also be an association between BBD and family history of breast cancer.[55]

An increased risk of breast cancer has also been demonstrated for women who have increased density of breast tissue as assessed by mammogram,[54,56,57] and breast density is likely to have a genetic component in its etiology.[58-60]

Other factors

Other risk factors, including those that are only weakly associated with breast cancer and those that have been inconsistently associated with the disease in epidemiologic studies (e.g., cigarette smoking), may be important in women who are in specific genotypically defined subgroups. One study [61] found a reduced risk of breast cancer among carriers of BRCA1/BRCA2 pathogenic variants who smoked, but an expanded follow-up study failed to find an association.[62]

Risk Factors for Ovarian Cancer

Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Prevention for information about risk factors for ovarian cancer in the general population.

Age

Ovarian cancer incidence rises in a linear fashion from age 30 years to age 50 years and continues to increase, though at a slower rate, thereafter. Before age 30 years, the risk of developing epithelial ovarian cancer is remote, even in hereditary cancer families.[63]

Family history including inherited cancer genes

Although reproductive, demographic, and lifestyle factors affect risk of ovarian cancer, the single greatest ovarian cancer risk factor is a family history of the disease. A large meta-analysis of 15 published studies estimated an odds ratio of 3.1 for the risk of ovarian cancer associated with at least one FDR with ovarian cancer.[64]

Reproductive history

Nulliparity is consistently associated with an increased risk of ovarian cancer, including among carriers of BRCA/BRCA2 pathogenic variants, yet a meta-analysis identified a risk reduction only in women with four or more live births.[15] Risk may also be increased among women who have used fertility drugs, especially those who remain nulligravid.[65,66] Several studies have reported a risk reduction in ovarian cancer after OC use in carriers of BRCA/BRCA2 pathogenic variants;[67-69] a risk reduction has also been shown after tubal ligation in BRCA1 carriers, with a statistically significant decreased risk of 22% to 80% after the procedure.[69,70] Breastfeeding for more than 12 months may also be associated with a reduction in ovarian cancer among carriers of BRCA1/BRCA2 pathogenic variants.[71] On the other hand, evidence is growing that the use of menopausal HRT is associated with an increased risk of ovarian cancer, particularly in long-time users and users of sequential estrogen-progesterone schedules.[72-75]

Surgical history

Bilateral tubal ligation and hysterectomy are associated with reduced ovarian cancer risk,[65,76,77] including in carriers of BRCA/BRCA2 pathogenic variants.[78] Ovarian cancer risk is reduced more than 90% in women with documented BRCA1 or BRCA2 pathogenic variants who chose risk-reducing salpingo-oophorectomy (RRSO). In this same population, risk-reducing oophorectomy also resulted in a nearly 50% reduction in the risk of subsequent breast cancer.[79,80] While some studies have shown more benefit for breast cancer reduction in patients with BRCA2 versus BRCA1 pathogenic variants, others have shown no benefit for BRCA1 carriers. Additionally, many of the studies remain underpowered to demonstrate benefit.[81] (Refer to the RRSO section of this summary for more information about these studies.)

Oral contraceptives

Use of OCs for 4 or more years is associated with an approximately 50% reduction in ovarian cancer risk in the general population.[65,82] A majority of, but not all, studies also support OCs being protective among carriers of BRCA/BRCA2 pathogenic variants.[70,83-86] A meta-analysis of 18 studies including 13,627 carriers of BRCA pathogenic variants reported a significantly reduced risk of ovarian cancer (SRR, 0.50; 95% CI, 0.33–0.75) associated with OC use.[20] (Refer to the Oral contraceptives section in the Chemoprevention section of this summary for more information.)

Risk Factors for Endometrial Cancer

Refer to the PDQ summary on Endometrial Cancer Prevention for information about risk factors for endometrial cancer in the general population.

Age

Age is an important risk factor for endometrial cancer. Most women with endometrial cancer are diagnosed after menopause. Only 15% of women are diagnosed with endometrial cancer before age 50 years, and fewer than 5% are diagnosed before age 40 years.[87] Women with Lynch syndrome tend to develop endometrial cancer at an earlier age, with the median age at diagnosis of 48 years.[88]

Family history including inherited cancer genes

Although the hyperestrogenic state is the most common predisposing factor for endometrial cancer, family history also plays a significant role in a woman’s risk for disease. Approximately 3% to 5% of uterine cancer cases are attributable to a hereditary cause,[89] with the main hereditary endometrial cancer syndrome being Lynch syndrome, an autosomal dominant genetic condition with a population prevalence of 1 in 300 to 1 in 1,000 individuals.[90,91] (Refer to the Lynch Syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information.)

Non-Lynch syndrome genes may also contribute to endometrial cancer risk. In an unselected endometrial cancer cohort undergoing multigene panel testing, approximately 3% of patients tested positive for a germline pathogenic variant in non-Lynch syndrome genes, including CHEK2, APC, ATM, BARD1, BRCA1, BRCA2, BRIP1, NBN, PTEN, and RAD51C.[92] Notably, patients with pathogenic variants in non-Lynch syndrome genes were more likely to have serous tumor histology than were patients without pathogenic variants. Furthermore, although the overall risk of endometrial cancer after RRSO was not increased among carriers of BRCA1 pathogenic variants, these patients seemed to have an increased risk of serous and serous-like endometrial cancer.[93] These findings were supported by a Dutch multicenter cohort study in women with germline BRCA1 and BRCA2 pathogenic variants. This study concluded that the absolute risk of endometrial cancer was approximately 3%. Because some serous and p53-aberrant endometrial cancers may harbor germline or somatic BRCA1/BRCA2 mutations, poly (ADP-ribose) polymerase (PARP) inhibitor therapy may also be a therapeutic option.[94]

Reproductive history

Reproductive factors such as multiparity, late menarche, and early menopause decrease the risk of endometrial cancer because of the lower cumulative exposure to estrogen and the higher relative exposure to progesterone.[95,96]

Hormones

Hormonal factors that increase the risk of type I endometrial cancer are better understood. All endometrial cancers share a predominance of estrogen relative to progesterone. Prolonged exposure to estrogen or unopposed estrogen increases the risk of endometrial cancer. Endogenous exposure to estrogen can result from obesity, polycystic ovary syndrome, and nulliparity, while exogenous estrogen can result from taking unopposed estrogen or tamoxifen. Unopposed estrogen increases the risk of developing endometrial cancer by twofold to twentyfold, proportional to the duration of use.[97,98] Tamoxifen, a selective estrogen receptor modulator, acts as an estrogen agonist on the endometrium while acting as an estrogen antagonist in breast tissue, and increases the risk of endometrial cancer.[99] In contrast, OCs, the levonorgestrel-releasing intrauterine system, and combination estrogen-progesterone hormone replacement therapy all reduce the risk of endometrial cancer through the antiproliferative effect of progesterone acting on the endometrium.[100-103]

Autosomal Dominant Inheritance of Breast and Gynecologic Cancer Predisposition

Autosomal dominant inheritance of breast and gynecologic cancers is characterized by transmission of cancer predisposition from generation to generation, through either the mother’s or the father’s side of the family, with the following characteristics:

  • Inheritance risk of 50%. When a parent carries an autosomal dominant genetic predisposition, each child has a 50:50 chance of inheriting the predisposition. Although the risk of inheriting the predisposition is 50%, not everyone with the predisposition will develop cancer because of incomplete penetrance and/or gender-restricted or gender-related expression.
  • Both males and females can inherit and transmit an autosomal dominant cancer predisposition. A male who inherits a cancer predisposition can still pass the altered gene on to his sons and daughters.

Breast and ovarian cancer are components of several autosomal dominant cancer syndromes. The syndromes most strongly associated with both cancers are the syndromes associated with BRCA1 or BRCA2 pathogenic variants. Breast cancer is also a common feature of Li-Fraumeni syndrome due to TP53 pathogenic variants and of Cowden syndrome due to PTEN pathogenic variants.[104] Other genetic syndromes that may include breast cancer as an associated feature include heterozygous carriers of the ataxia telangiectasia gene and Peutz-Jeghers syndrome. Ovarian cancer has also been associated with Lynch syndrome, basal cell nevus (Gorlin) syndrome, and multiple endocrine neoplasia type 1.[104] Lynch syndrome is mainly associated with colorectal cancer and endometrial cancer, although several studies have demonstrated that patients with Lynch syndrome are also at risk of developing transitional cell carcinoma of the ureters and renal pelvis; cancers of the stomach, small intestine, liver and biliary tract, brain, breast, prostate, and adrenal cortex; and sebaceous skin tumors (Muir-Torre syndrome).[105-111]

Germline pathogenic variants in the genes responsible for these autosomal dominant cancer syndromes produce different clinical phenotypes of characteristic malignancies and, in some instances, associated nonmalignant abnormalities.

The family characteristics that suggest hereditary cancer predisposition include the following:

  • Multiple cancers within a family.
  • Cancers typically occur at an earlier age than in sporadic cases (defined as cases not associated with genetic risk).
  • Two or more primary cancers in a single individual. These could be multiple primary cancers of the same type (e.g., bilateral breast cancer) or primary cancer of different types (e.g., breast cancer and ovarian cancer in the same individual or endometrial and colon cancer in the same individual).
  • Cases of male breast cancer. The inheritance risk for autosomal dominant genetic conditions is 50% for both males and females, but the differing penetrance of the genes may result in some unaffected individuals in the family.

Figure 1 and Figure 2 depict some of the classic inheritance features of a BRCA1 and BRCA2 pathogenic variant, respectively. Figure 3 depicts a classic family with Lynch syndrome. (Refer to the Standard Pedigree Nomenclature figure in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for definitions of the standard symbols used in these pedigrees.)

Enlarge Pedigree showing some of the classic features of a family with a deleterious BRCA1 mutation across three generations, including transmission occurring through maternal and paternal lineages. The unaffected female proband is shown as having an affected mother (breast cancer diagnosed at age 42 y), female cousin (breast cancer diagnosed at age 38 y), maternal aunt (ovarian cancer diagnosed at age 53 y), and maternal grandmother (ovarian cancer diagnosed at age 49 y).
Figure 1. BRCA1 pedigree. This pedigree shows some of the classic features of a family with a BRCA1 pathogenic variant across three generations, including affected family members with breast cancer or ovarian cancer and a young age at onset. BRCA1 families may exhibit some or all of these features. As an autosomal dominant syndrome, a BRCA1 pathogenic variant can be transmitted through maternal or paternal lineages, as depicted in the figure.
Enlarge Pedigree showing some of the classic features of a family with a deleterious BRCA2 mutation across three generations, including transmission occurring through maternal and paternal lineages. The unaffected female proband is shown as having an affected brother (breast cancer diagnosed at age 52 y), mother (breast cancer diagnosed at age 45 y and pancreatic cancer diagnosed at age 55 y), maternal aunt (ovarian cancer diagnosed at age 58 y), and maternal grandfather (prostate cancer diagnosed at age 55 y).
Figure 2. BRCA2 pedigree. This pedigree shows some of the classic features of a family with a BRCA2 pathogenic variant across three generations, including affected family members with breast (including male breast cancer), ovarian, pancreatic, or prostate cancers and a relatively young age at onset. BRCA2 families may exhibit some or all of these features. As an autosomal dominant syndrome, a BRCA2 pathogenic variant can be transmitted through maternal or paternal lineages, as depicted in the figure.
Enlarge Pedigree showing some of the classic features of a family with Lynch syndrome across three generations, including transmission occurring through maternal and paternal lineages and the presence of both colon and endometrial cancers.
Figure 3. Lynch syndrome pedigree. This pedigree shows some of the classic features of a family with Lynch syndrome, including affected family members with colon cancer or endometrial cancer, a young age at onset in some individuals, and incomplete penetrance. Lynch syndrome families may exhibit some or all of these features. Lynch syndrome families may also include individuals with other gastrointestinal, gynecologic, and genitourinary cancers, or other extracolonic cancers. As an autosomal dominant syndrome, Lynch syndrome can be transmitted through maternal or paternal lineages, as depicted in the figure. Because the cancer risk is not 100%, individuals who have Lynch syndrome may not develop cancer, such as the mother of the female with colon cancer diagnosed at age 37 years in this pedigree (called incomplete penetrance).

There are no pathognomonic features distinguishing breast and ovarian cancers occurring in carriers of BRCA1 or BRCA2 pathogenic variants from those occurring in noncarriers. Breast cancers occurring in carriers of BRCA1 pathogenic variants are more likely to be ER-negative, progesterone receptor–negative, HER2/neu receptor–negative (i.e., triple-negative breast cancers [TNBC]), and have a basal phenotype. BRCA1-associated ovarian cancers are more likely to be high-grade and of serous histopathology. (Refer to the Pathology of breast cancer and Pathology of ovarian cancer sections of this summary for more information.)

Some pathologic features distinguish carriers of Lynch syndrome–associated pathogenic variants from noncarriers. The hallmark feature of endometrial cancers occurring in Lynch syndrome is mismatch repair (MMR) deficiencies, including the presence of microsatellite instability (MSI), and the absence of specific MMR proteins. In addition to these molecular changes, there are also histologic changes including tumor-infiltrating lymphocytes, peritumoral lymphocytes, undifferentiated tumor histology, lower uterine segment origin, and synchronous tumors.

Considerations in Risk Assessment and in Identifying a Family History of Breast and Ovarian Cancer Risk

The accuracy and completeness of family histories must be considered when they are used to assess risk. A reported family history may be erroneous, or a person may be unaware of relatives affected with cancer. In addition, small family sizes and premature deaths may limit the information obtained from a family history. Breast or ovarian cancer on the paternal side of the family usually involves more distant relatives than does breast or ovarian cancer on the maternal side, so information may be more difficult to obtain. When self-reported information is compared with independently verified cases, the sensitivity of a history of breast cancer is relatively high, at 83% to 97%, but lower for ovarian cancer, at 60%.[112,113] Additional limitations of relying on family histories include adoption; families with a small number of women; limited access to family history information; and incidental removal of the uterus, ovaries, and/or fallopian tubes for noncancer indications. Family histories will evolve; therefore, it is important to update family histories from both parents over time. (Refer to the Accuracy of the family history section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)

Models for Prediction of Breast and Gynecologic Cancer Risk

Models to predict an individual’s lifetime risk of developing breast and/or gynecologic cancer are available.[114-117] In addition, models exist to predict an individual’s likelihood of having a pathogenic variant in BRCA1, BRCA2, or one of the MMR genes associated with Lynch syndrome. (Refer to the Models for prediction of the likelihood of a BRCA1 or BRCA2 pathogenic variant section of this summary for more information about some of these models.) Not all models can be appropriately applied to all patients. Each model is appropriate only when the patient’s characteristics and family history are similar to those of the study population on which the model was based. Different models may provide widely varying risk estimates for the same clinical scenario, and the validation of these estimates has not been performed for many models.[115,118,119]

Breast cancer risk assessment models

In general, breast cancer risk assessment models are designed for two types of populations: 1) women without a pathogenic variant or strong family history of breast or ovarian cancer; and 2) women at higher risk because of a personal or family history of breast cancer or ovarian cancer.[119] Models designed for women of the first type (e.g., the Gail model, which is the basis for the Breast Cancer Risk Assessment Tool [BCRAT]) [120], and the Colditz and Rosner model [121]) require only limited information about family history (e.g., number of FDRs with breast cancer). Models designed for women at higher risk require more detailed information about personal and family cancer history of breast and ovarian cancers, including ages at onset of cancer and/or carrier status of specific breast cancer-susceptibility alleles. The genetic factors used by the latter models differ, with some assuming one risk locus (e.g., the Claus model [122]), others assuming two loci (e.g., the International Breast Cancer Intervention Study [IBIS] model [123] and the BRCAPRO model [124]), and still others assuming an additional polygenic component in addition to multiple loci (e.g., the Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm [BOADICEA] model [125-127]). The models also differ in whether they include information about nongenetic risk factors. Three models (Gail/BCRAT, Pfeiffer,[117] and IBIS) include nongenetic risk factors but differ in the risk factors they include (e.g., the Pfeiffer model includes alcohol consumption, whereas the Gail/BCRAT does not). These models have limited ability to discriminate between individuals who are affected and those who are unaffected with cancer; a model with high discrimination would be close to 1, and a model with little discrimination would be close to 0.5; the discrimination of the models currently ranges between 0.56 and 0.63).[128] The existing models generally are more accurate in prospective studies that have assessed how well they predict future cancers.[119,129-131] An analysis comparing the 10-year performance of the BOADICEA, BRCAPRO, BCRAT, and IBIS models demonstrated superiority of the models with more detailed pedigree inclusion—specifically, BOADICEA and IBIS.[132]

In the United States, BRCAPRO, the Claus model,[122,133] and the Gail/BCRAT [120] are widely used in clinical counseling. Risk estimates derived from the models differ for an individual patient. Several other models that include more detailed family history information are also in use and are discussed below.

Additional considerations for clinical use of breast cancer risk assessment models

The Gail model is the basis for the BCRAT, a computer program available from the National Cancer Institute (NCI) by calling the Cancer Information Service at 1-800-4-CANCER (1-800-422-6237). This version of the Gail model estimates only the risk of invasive breast cancer. The Gail/BCRAT model has been found to be reasonably accurate at predicting breast cancer risk in large groups of White women who undergo annual screening mammography; however, reliability varies depending on the cohort studied.[134-139] Risk can be overestimated in the following populations:

  • Women who do not adhere to screening recommendations.[134,135]
  • Women in the highest-risk strata.[137]

The Gail/BCRAT model is valid for women aged 35 years and older. The model was primarily developed for White women.[138] Extensions of the Gail model for African American women have been subsequently developed to calibrate risk estimates using data from more than 1,600 African American women with invasive breast cancer and more than 1,600 controls.[140] Additionally, extensions of the Gail model have incorporated high-risk single nucleotide variants and pathogenic variants; however, no software exists to calculate risk in these extended models.[141,142] Other risk assessment models incorporating breast density have been developed but are not ready for clinical use.[143,144]

Generally, the Gail/BCRAT model should not be the sole model used for families with one or more of the following characteristics:

  • Multiple affected individuals with breast cancer or ovarian cancer (especially when one or more breast cancers are diagnosed before age 50 y).
  • A woman with both breast and ovarian cancer.
  • Ashkenazi Jewish ancestry with at least one case of breast or ovarian cancer (as these families are more likely to have a hereditary cancer susceptibility syndrome).

Commonly used models that incorporate family history include the IBIS, BOADICEA, and BRCAPRO models. The IBIS/Tyrer-Cuzick model incorporates both genetic and nongenetic factors.[123] A three-generation pedigree is used to estimate the likelihood that an individual carries either a BRCA1/BRCA2 pathogenic variant or a hypothetical low-penetrance gene. In addition, the model incorporates personal risk factors such as parity, body mass index (BMI); height; and age at menarche, first live birth, menopause, and HRT use. Both genetic and nongenetic factors are combined to develop a risk estimate. The BOADICEA model examines family history to estimate breast cancer risk and also incorporates both BRCA1/BRCA2 and non-BRCA1/BRCA2 genetic risk factors.[126] The most important difference between BOADICEA and the other models using information on BRCA1/BRCA2 is that BOADICEA assumes an additional polygenic component in addition to multiple loci,[125-127] which is more in line with what is known about the underlying genetics of breast cancer. The BOADICEA model has also been expanded to include additional pathogenic variants, including CHEK2, ATM, and PALB2.[145] However, the discrimination and calibration for these models differ significantly when compared in independent samples;[129] the IBIS and BOADICEA models are more comparable when estimating risk over a shorter fixed time horizon (e.g., 10 years),[129] than when estimating remaining lifetime risk. As all risk assessment models for cancers are typically validated over a shorter time horizon (e.g., 5 or 10 years), fixed time horizon estimates rather than remaining lifetime risk may be more accurate and useful measures to convey in a clinical setting.

In addition, readily available models that provide information about an individual woman’s risk in relation to the population-level risk depending on her risk factors may be useful in a clinical setting (e.g., Your Disease Risk). Although this tool was developed using information about average-risk women and does not calculate absolute risk estimates, it still may be useful when counseling women about prevention. Risk assessment models are being developed and validated in large cohorts to integrate genetic and nongenetic data, breast density, and other biomarkers.

Although most breast cancer risk models have been shown to be well calibrated overall, model performance can be different for subgroups of women. In particular, independent, prospective validation of risk models for women who tested negative for BRCA1 or BRCA2 pathogenic variants supported that the most commonly used clinical risk models underpredicted risk for this group of women.[146] The performance also differed on the basis of whether the test results of relatives were known. The models also underpredicted risk by 26.3% to 56.7% in women who tested negative but whose relatives had not been tested.

Ovarian cancer risk assessment models

Two risk prediction models have been developed for ovarian cancer.[116,117] The Rosner model [116] included age at menopause, age at menarche, oral contraception use, and tubal ligation; the concordance statistic was 0.60 (0.57–0.62). The Pfeiffer model [117] included oral contraceptive use, menopausal hormone therapy use, and family history of breast cancer or ovarian cancer, with a similar discriminatory power of 0.59 (0.56–0.62). Although both models were well calibrated, their modest discriminatory power limited their screening potential.

Endometrial cancer risk assessment models

The Pfeiffer model has been used to predict endometrial cancer risk in the general population.[117] For endometrial cancer, the relative risk model included BMI, menopausal hormone therapy use, menopausal status, age at menopause, smoking status, and OC use. The discriminatory power of the model was 0.68 (0.66–0.70); it overestimated observed endometrial cancers in most subgroups but underestimated disease in women with the highest BMI category, in premenopausal women, and in women taking menopausal hormone therapy for 10 years or more.

In contrast, MMRpredict, PREMM5 (PREdiction Model for gene Mutations), and MMRpro are three quantitative predictive models used to identify individuals who may potentially have Lynch syndrome.[147-149] MMRpredict incorporates only colorectal cancer patients but does include MSI and immunohistochemistry (IHC) tumor testing results. PREMM5 is an update of PREMM(1,2,6) and includes each of the five genes associated with Lynch syndrome, including PMS2 and EPCAM. It accounts for other Lynch syndrome–associated tumors but does not include tumor testing results.[148] MMRpro incorporates tumor testing and germline testing results, but is more time intensive because it includes affected and unaffected individuals in the risk-quantification process. All three predictive models are comparable to the traditional Amsterdam and Bethesda criteria in identifying individuals with colorectal cancer who carry MMR gene pathogenic variants.[150] However, because these models were developed and validated in colorectal cancer patients, the discriminative abilities of these models to identify Lynch syndrome are lower among individuals with endometrial cancer than among those with colon cancer.[151] In fact, the sensitivity and specificity of MSI and IHC in identifying carriers of pathogenic variants are considerably higher than the prediction models and support the use of molecular tumor testing to screen for Lynch syndrome in women with endometrial cancer.

Table 1 summarizes salient aspects of breast and gynecologic cancer risk assessment models that are commonly used in the clinical setting. These models differ by the extent of family history included, whether nongenetic risk factors are included, and whether carrier status and polygenic risk are included (inputs to the models). The models also differ in the type of risk estimates that are generated (outputs of the models). These factors may be relevant in choosing the model that best applies to a particular individual.

Table 1. Summary of Prediction Models Used to Calculate Age-Specific Absolute Risks of Breast and Gynecologic Cancers
Model Family History (input) Pathogenic Variants (input) Risk Factors (input) Risk Estimate Generated (output)
BCRAT = Breast Cancer Risk Assessment Tool; BOADICEA = Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm; IBIS = International Breast Cancer Intervention Study; PREMM = PREdiction Model for gene Mutations.
aHigh risk is defined as those with a personal or family history of the designated cancer type.
bTakes into account polygenes as an underlying assumption of the model.
Breast Cancer Risk Assessment Models
Models for Average-Risk Women
Gail/BCRAT First-degree relatives (breast cancer) No Yes Breast cancer
Pfeiffer (breast) [117] First-degree relatives (breast, ovarian cancers) No Yes Breast cancer
Colditz and Rosner [121] None No Yes Breast cancer
Models for High-Risk Womena
Claus [122] Multigenerational (breast cancer) No No Breast cancer
BRCAPRO Multigenerational (breast, ovarian cancers) BRCA1/BRCA2 No Breast cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant
IBIS Multigenerational (ovarian cancer) BRCA1/BRCA2 Yes Breast cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant
BOADICEAb Multigenerational (pancreatic, breast, ovarian cancers) BRCA1/BRCA2 No Breast and ovarian cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant
Ovarian Cancer Risk Assessment Models
Models for Average-Risk Women
Rosner [116] None No Yes Ovarian cancer
Pfeiffer (ovarian) [117] First-degree relatives (breast, ovarian cancers) No Yes Breast cancer
Models for High-Risk Womena
BOADICEAb Multigenerational (pancreatic, breast, ovarian cancers) BRCA1/BRCA2 No Breast and ovarian cancer; % risk of carrying BRCA1/BRCA2 pathogenic variant
Endometrial Cancer Risk Assessment Models
Models for Average-Risk Women
Pfeiffer (endometrial) [117] None No Yes Endometrial cancer
Models for High-Risk Womena
PREMM5 Multigenerational (colon, endometrial and other Lynch syndrome–associated cancers and polyps) No No % risk of carrying MLH1, MSH2, MSH6 pathogenic variant
MMRpro Multigenerational (colon, endometrial cancers) No No % risk of carrying MLH1, MSH2, MSH6 pathogenic variant
MMRpredict [147] Multigenerational (colon, endometrial cancers) No No % risk of carrying MLH1, MSH2, MSH6 pathogenic variant

Considerations When Conducting Genetic Testing

Indications for genetic testing

Several professional organizations and expert panels— including the American Society of Clinical Oncology,[152] the National Comprehensive Cancer Network (NCCN),[153] the American Society of Human Genetics,[154] the American College of Medical Genetics and Genomics,[155] the National Society of Genetic Counselors,[155] the U.S. Preventive Services Task Force,[156] and the Society of Gynecologic Oncologists —[157] have developed clinical criteria and practice guidelines that can be helpful to health care providers in identifying individuals who may have a BRCA1 or BRCA2 pathogenic variant.

In 2019, the American Society of Breast Surgeons published a recommendation to make genetic testing for “BRCA1/BRCA2, and PALB2, with other genes as appropriate for the clinical scenario and family history” available to all breast cancer patients.[158] This recommendation was based on a study that suggested similar pathogenic variant rates identified through an extended multigene panel in patients with breast cancer who did or did not meet the NCCN guidelines for genetic testing.[159] This study had important methodologic challenges that need to be considered, including exclusion of participants previously tested, uncertain accuracy of the reported risk criteria for study participants, inclusion of genes with uncertain management guidelines, and difference in the specific genes in which pathogenic or likely pathogenic variants were identified across the two groups. For example, there was a statistically significant difference between participants who met and who did not meet NCCN criteria in the detection of BRCA1/BRCA2 variants.

Other studies have also found that the NCCN criteria have good sensitivity when predicting BRCA1/BRCA2 variants; however, less is known about many other genes. For example, one study showed that the NCCN criteria were able to detect 88.9% of the BRCA1/BRCA2 pathogenic variant carriers [160] and others have found that, if more than one NCCN criterion is met, then the positive predictive value does pass the 10% threshold (e.g., 12% for more than two NCCN criteria).[161]

As the cost of genetic testing continues to decrease, there is a need for unbiased evidence to guide indications for testing, including the cost-benefit impact on screening, prevention, and treatment. Efforts to generate less biased evidence include a single institution study of 3,907 unselected women with breast cancer tested for nine breast cancer genes, including BRCA1/BRCA2, ATM, CDH1, CHEK2, NF1, PALB2, PTEN, and TP53.[162] The study assessed the relative performance of NCCN genetic testing criteria as compared with the American Society of Breast Surgeons' recommendation to test all women aged 65 years or younger with breast cancer. The sensitivity of the criteria was defined as the proportion of individuals who met testing criteria and tested positive for a pathogenic or likely pathogenic variant of the total population of pathogenic or likely pathogenic variant carriers in the study, while the specificity was defined as the proportion of individuals who did not meet testing criteria and tested negative for a pathogenic or likely pathogenic variant of the total population of noncarriers in the study. High sensitivity and specificity are both important considerations; however, higher sensitivity leads to lower specificity, so it is important to balance these two factors. Detection of BRCA1/BRCA2 pathogenic or likely pathogenic variants based on NCCN criteria had a sensitivity of 87% with a specificity of 53.5%; when expanded to the nine genes included in the study, sensitivity was 70% and specificity was 53.2%. When including all women diagnosed with breast cancer at age 65 or younger, the sensitivity to detect BRCA1/BRCA2 pathogenic or likely pathogenic variants increased to 98%, while the specificity dropped to 22%. Among those who did not meet NCCN criteria, 0.7% had pathogenic or likely pathogenic BRCA1/BRCA2 variants.

Another study to assess frequency of pathogenic or likely pathogenic variants among breast cancer patients included a nested case-control study conducted through the Women’s Health Initiative (WHI) cohort among women with (cases) and without (controls) invasive breast cancer. Participants were tested for pathogenic or likely pathogenic variants in ten breast cancer–associated genes, including BRCA1/BRCA2.[163] The prevalence of pathogenic or likely pathogenic BRCA1/BRCA2 variants among those diagnosed with invasive breast cancer before age 65 years was 2.21%, compared with 1.09% among those diagnosed at age 65 years or older. In comparison, the frequency of pathogenic or likely pathogenic BRCA1/BRCA2 variants was 0.22% in the control group. Current genetic testing criteria detect BRCA pathogenic variants. Although higher sensitivity is always desired, it is at the expense of specificity. Lower specificity leads to higher costs to achieve one positive genetic test.

Benefits of offering genetic testing at the time of cancer diagnosis

At the time of a new cancer diagnosis, genetic testing for inherited cancer predisposition may guide patient care including decisions about surgery, chemotherapy and other biologics, and radiation treatment.[164,165] Among high-risk patients, the option of genetic testing is an important part of the shared decision-making process regarding cancer treatments at the time of diagnosis. Tools are available to facilitate decision making about genetic testing in this context.[166]

Breast cancer diagnosis

Benefits of offering genetic testing at the time of breast cancer diagnosis include, but are not limited to, the following:

  1. Surgery: The identification of inherited susceptibility to breast cancer may influence surgical treatment decisions. As an example, the high risk of a second primary breast cancer among BRCA pathogenic variant carriers, particularly those diagnosed at an early age, may influence their decision to choose a bilateral mastectomy (versus a lumpectomy or unilateral/subtotal mastectomy) for surgical treatment of their breast cancer.[167] (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section of this summary for more information about the risk of a second primary breast cancer.) Discussion of risk-reducing salpingo-oophorectomy is indicated,[168] and referral to a gynecologic provider may be considered.
  2. Chemotherapy and other biologics: Medical treatments may be guided by the identification of a pathogenic variant in an inherited cancer predisposing gene. As an example, among BRCA pathogenic variant carriers, breast cancer treatment may include the use of platinum-based agents.[169] Furthermore, novel agents such as PARP inhibitors may be used in the treatment of metastatic breast cancer.[170]
  3. Radiation therapy: Decisions about the use of radiation treatment may be guided by the presence of a pathogenic variant in an inherited breast cancer susceptibility gene. In particular, the poorer wound healing in irradiated breasts is an important consideration for those who may consider risk-reducing mastectomy with reconstruction. As an example, individuals with a pathogenic variant in TP53 may experience higher risks from radiation, including increased risks for subsequent new cancers.[171,172] Thus, identification of TP53 carriers in the context of an active breast cancer diagnosis may influence radiation treatment decisions and reconstruction options.
Ovarian cancer diagnosis

Benefits of offering genetic testing at the time of ovarian cancer diagnosis include, but are not limited to, the following:

  1. Surgery: In most cases, the decision for ovarian cancer surgery is made on the basis of an adnexal mass or abdominal symptoms. When possible, considering the likelihood of a heritable genetic variant at the time of diagnosis may add value to surgical decision-making. The identification of inherited susceptibility to ovarian/fallopian tube cancer may influence surgical treatment decisions. For a questionable adnexal mass in a younger woman who is at risk of carrying a pathogenic variant of a highly penetrant ovarian cancer gene, knowledge of this information may help guide a decision for risk-reducing or therapeutic surgery.[173,174] For women who may be considering fertility preservation surgery, genetic knowledge may motivate consideration of bilateral salpingo-oophorectomy, and in the case of carriers of BRCA1 pathogenic variants, a more detailed discussion regarding aggressive uterine cancer risk.
  2. Chemotherapy and other biologics: First-line chemotherapy for ovarian cancer still relies on a backbone of platinum and taxane chemotherapy. Current treatment options for optimally resected stage III ovarian carcinoma include intravenous (IV) chemotherapy, dose-dense IV chemotherapy, and a combination of IV paclitaxel plus intraperitoneal (IP) cisplatin, followed by IP paclitaxel 1 week later. Carriers of BRCA1 and BRCA2 pathogenic variants are considered more platinum sensitive, with longer progression-free survival times compared with BRCA1 and BRCA2 wild-type patients,[175,176] so it is unclear whether a particular treatment strategy is driven more by antiangiogenesis effects, peritoneal dose intensity, or platinum dose intensity. The advent of PARP as a biologic target (in combination with chemotherapy or as maintenance) may also increase the armory of first-line treatment of ovarian cancer.[177] (Refer to the Systemic therapy in ovarian cancer treatment section in the Ovarian cancer section of this summary for more information about PARP inhibitors in ovarian cancer treatment.)
Endometrial cancer diagnosis

Benefits of offering genetic testing at the time of endometrial cancer diagnosis include, but are not limited to, the following:

  1. Surgery: The most common treatment for a newly diagnosed endometrial cancer includes hysterectomy with removal of the ovaries and fallopian tubes, as well as assessment of lymph nodes.[178] An exception to this practice might apply to a younger woman who wishes to retain fertility or retain her adnexa. Immunohistochemistry of endometrial sampling may allow for an assessment of the likelihood of a heritable genetic variant at the time of diagnosis, which may add value to the surgical decision-making process. For a young woman who is found to have Lynch syndrome, knowledge of this information may help guide a decision for hormonal management of endometrial cancer to allow future childbearing, or salpingo-oophorectomy if her risk of ovarian cancer is deemed high enough on the basis of a specific genetic variant. For a young woman who is found to carry a pathogenic variant in BRCA1/BRCA2, or one of the other homologous recombination deficiencies increasing ovarian cancer risk, she may wish to decide between salpingo-oophorectomy or, at least, salpingectomy.
  2. Chemotherapy and other biologics: Immune checkpoint inhibitors are now approved for use in endometrial cancers that have MSI or MMR deficiency.[179] While MSI and MMR status can be assessed at either the time of diagnosis or recurrent disease, it may be beneficial to perform tumor testing at diagnosis with the primary pathology processing, usually at the time of hysterectomy.

Multigene (panel) testing

Since the availability of next-generation sequencing and the Supreme Court of the United States ruling that human genes cannot be patented, several clinical laboratories now offer genetic testing through multigene panels at a cost comparable to that of single-gene testing. Even testing for BRCA1 and BRCA2 is a limited panel test of two genes. Approximately 25% of all ovarian/fallopian tube/peritoneal cancers are caused by a heritable genetic condition. Of these, about one-quarter (6% of all ovarian/fallopian tube/peritoneal cancers) are caused by genes other than BRCA1 and BRCA2, including many genes associated with the Fanconi anemia pathway or otherwise involved with homologous recombination.[180] In a population of ovarian cancer patients who test negative for BRCA1 and BRCA2 pathogenic variants, multigene panel testing can reveal actionable pathogenic variants.[181-183]

In general, multigene panel testing increases the yield of non-BRCA pathogenic variants across a variety of populations.[165,184-186] In an unselected population of breast cancer patients, the prevalence of BRCA1 and BRCA2 pathogenic variants was 6.1%, while the prevalence of pathogenic variants in other breast/ovarian cancer–predisposing genes was 4.6%.[187] In an unselected population of endometrial cancer patients, the prevalence of Lynch syndrome pathogenic variants (MLH1, MSH2, EPCAM-MSH2, MSH6, and PMS2) was 5.8%; the prevalence of pathogenic variants in other actionable genes was 3.4%.[92] Similarly, in a study of 35,409 women with breast cancer tested with the Myriad 25-gene panel, a pathogenic variant was found in 9.3% of women.[188] Among that 9.3%, 48.5% of the women carried a pathogenic variant in BRCA1 or BRCA2. The majority of other breast cancer genes with pathogenic variants identified included CHEK2 (11.7%), ATM (9.7%), and PALB2 (9.3%). The prevalence of pathogenic variants in the other breast cancer genes on the panel ranged from 0.05% to 0.31%. Pathogenic variants in Lynch syndrome genes accounted for 7.0% of variants identified; 3.7% were found in other genes included in the panel. The rate of pathogenic variants was higher in women with TNBC diagnosed before age 40 years. A similar trend of identifying pathogenic variants in non-BRCA susceptibility genes in male breast cancer patients has also been described.[189] In two studies of women who had previously tested negative for BRCA1/BRCA2, reflex testing with a multigene panel identified pathogenic variants in additional genes among 8% to 11% of cases.[190,191] In a study of 77,085 patients with breast cancer and 6,001 patients with ovarian cancer, 24.1% and 30.9% had genetic testing, respectively. Of those tested, pathogenic or likely pathogenic variants were identified in 7.8% of patients with breast cancer and 14.5% of patients with ovarian cancer. Prevalent non-BRCA pathogenic variants identified in patients with breast cancer included CHEK2 (1.6%), PALB2 (1.0%), ATM (0.7%), and NBN (0.4%). In patients with ovarian cancer, non-BRCA pathogenic variants included CHEK2 (1.4%), BRIP1 (0.9%), MSH2 (0.8%), and ATM (0.6%).[192] The potential utility of genetic testing in patients with ovarian tumors of all histologies was suggested in a study using a 32-gene panel that found 13.2% of 4,439 tumors harbored a pathogenic variant. Rates were highest among those with serous ovarian carcinoma (14.7%), although likely pathogenic variants were also seen in those with other histologies (borderline, germ cell, and sex cord stromal tumors), the significance of which is unclear to clinical management or etiology of disease.[193]

Women diagnosed with TNBC are currently recommended to undergo BRCA1/BRCA2 testing to guide treatment decisions at any age.[153] A large study utilizing multigene (panel) testing comprising two separate cohorts reported that, in addition to BRCA1/BRCA2 genes, six other breast cancer susceptibility genes were also related to a higher risk of TNBC. Specifically, pathogenic variants in BARD1, PALB2, and RAD51D, in addition to BRCA1 and BRCA2, were each associated with more than a fivefold increase in breast cancer.[194] Pathogenic variants in three other genes —BRIP1, RAD51C, and TP53— were each associated with an increased TNBC risk of more than twofold. Pathogenic variants in these eight genes were reported in 12% of the TNBC cases (8.3% BRCA1/BRCA2, 3.7% non-BRCA1/BRCA2). The study was conducted in a clinical testing cohort of 140,449 individuals (8,753 TNBC cases) who received genetic testing using a 21-gene panel (sample A). In addition, a second sample (sample B) examined gene frequency rates in a pooled consortium of 2,143 individuals using a 17-gene panel. The overall frequency of pathogenic variants in the 21 genes examined in sample A was 14.4% (8.4% BRCA1/BRCA2, 6.0% non-BRCA1/BRCA2). The two samples had very consistent findings with respect to the risk estimates despite differences in age, race, ethnicity, and family history of cancer with sample A being younger, more racially and ethnically diverse, and more likely to have a family history of cancer. The pathogenic variant frequency detection in these 21 genes was also similar for White individuals (14% overall, 7.8% BRCA1/BRCA2, 6.2% non-BRCA1/BRCA2) and African American individuals (14.6% overall, 9.0% BRCA1/BRCA2, 5.6% non-BRCA1/BRCA2) supporting that the higher rates of TNBC in African American individuals versus White individuals is driven by environmental factors.

There are caveats of multigene testing. Genes identified as part of multigene panel testing can be associated with varied breast cancer risk or confer no known risk.[183] There is also the possibility of finding a variant of uncertain significance (VUS). Even within a given gene, there may be differential risks on the basis of specific pathogenic variants.[195] A large population-based retrospective study using Surveillance, Epidemiology, and End Results (SEER) program data from Georgia and Los Angeles, California, found that multigene testing led to a twofold increase in the detection of pathogenic variants compared with BRCA-only testing in women with breast cancer.[196] VUS rates, however, were tenfold higher in the multigene panels, especially in African American women (44.5%) and Asian women (50.9%). Many centers now offer a multigene panel test instead of just BRCA1 and BRCA2 testing if there is a concerning family history of syndromes other than hereditary breast and ovarian cancer, or more importantly, to gain as much genetic information as possible with one test, particularly if there may be insurance limitations.

(Refer to the Multigene [panel] testing section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information about multigene testing, including genetic education and counseling considerations and research examining the use of multigene testing.)

References
  1. American Cancer Society: Cancer Facts and Figures 2022. American Cancer Society, 2022. Available online. Last accessed January 12, 2022.
  2. Ravdin PM, Cronin KA, Howlader N, et al.: The decrease in breast-cancer incidence in 2003 in the United States. N Engl J Med 356 (16): 1670-4, 2007. [PUBMED Abstract]
  3. Feuer EJ, Wun LM, Boring CC, et al.: The lifetime risk of developing breast cancer. J Natl Cancer Inst 85 (11): 892-7, 1993. [PUBMED Abstract]
  4. Yang Q, Khoury MJ, Rodriguez C, et al.: Family history score as a predictor of breast cancer mortality: prospective data from the Cancer Prevention Study II, United States, 1982-1991. Am J Epidemiol 147 (7): 652-9, 1998. [PUBMED Abstract]
  5. Colditz GA, Willett WC, Hunter DJ, et al.: Family history, age, and risk of breast cancer. Prospective data from the Nurses' Health Study. JAMA 270 (3): 338-43, 1993. [PUBMED Abstract]
  6. Slattery ML, Kerber RA: A comprehensive evaluation of family history and breast cancer risk. The Utah Population Database. JAMA 270 (13): 1563-8, 1993. [PUBMED Abstract]
  7. Johnson N, Lancaster T, Fuller A, et al.: The prevalence of a family history of cancer in general practice. Fam Pract 12 (3): 287-9, 1995. [PUBMED Abstract]
  8. Pharoah PD, Day NE, Duffy S, et al.: Family history and the risk of breast cancer: a systematic review and meta-analysis. Int J Cancer 71 (5): 800-9, 1997. [PUBMED Abstract]
  9. Bevier M, Sundquist K, Hemminki K: Risk of breast cancer in families of multiple affected women and men. Breast Cancer Res Treat 132 (2): 723-8, 2012. [PUBMED Abstract]
  10. Kharazmi E, Chen T, Narod S, et al.: Effect of multiplicity, laterality, and age at onset of breast cancer on familial risk of breast cancer: a nationwide prospective cohort study. Breast Cancer Res Treat 144 (1): 185-92, 2014. [PUBMED Abstract]
  11. Reiner AS, Sisti J, John EM, et al.: Breast Cancer Family History and Contralateral Breast Cancer Risk in Young Women: An Update From the Women's Environmental Cancer and Radiation Epidemiology Study. J Clin Oncol 36 (15): 1513-1520, 2018. [PUBMED Abstract]
  12. Mucci LA, Hjelmborg JB, Harris JR, et al.: Familial Risk and Heritability of Cancer Among Twins in Nordic Countries. JAMA 315 (1): 68-76, 2016. [PUBMED Abstract]
  13. Johannsson O, Loman N, Borg A, et al.: Pregnancy-associated breast cancer in BRCA1 and BRCA2 germline mutation carriers. Lancet 352 (9137): 1359-60, 1998. [PUBMED Abstract]
  14. Jernström H, Lerman C, Ghadirian P, et al.: Pregnancy and risk of early breast cancer in carriers of BRCA1 and BRCA2. Lancet 354 (9193): 1846-50, 1999. [PUBMED Abstract]
  15. 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 106 (6): dju091, 2014. [PUBMED Abstract]
  16. Jernström H, Lubinski J, Lynch HT, et al.: Breast-feeding and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 96 (14): 1094-8, 2004. [PUBMED Abstract]
  17. Valentini A, Lubinski J, Byrski T, et al.: The impact of pregnancy on breast cancer survival in women who carry a BRCA1 or BRCA2 mutation. Breast Cancer Res Treat 142 (1): 177-85, 2013. [PUBMED Abstract]
  18. Milne RL, Osorio A, Ramón y Cajal T, et al.: Parity and the risk of breast and ovarian cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 119 (1): 221-32, 2010. [PUBMED Abstract]
  19. Breast cancer and hormonal contraceptives: collaborative reanalysis of individual data on 53 297 women with breast cancer and 100 239 women without breast cancer from 54 epidemiological studies. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet 347 (9017): 1713-27, 1996. [PUBMED Abstract]
  20. 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 46 (12): 2275-84, 2010. [PUBMED Abstract]
  21. Breast cancer and hormone replacement therapy: collaborative reanalysis of data from 51 epidemiological studies of 52,705 women with breast cancer and 108,411 women without breast cancer. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet 350 (9084): 1047-59, 1997. [PUBMED Abstract]
  22. Writing Group for the Women's Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 288 (3): 321-33, 2002. [PUBMED Abstract]
  23. Chlebowski RT, Hendrix SL, Langer RD, et al.: Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. JAMA 289 (24): 3243-53, 2003. [PUBMED Abstract]
  24. Beral V; Million Women Study Collaborators: Breast cancer and hormone-replacement therapy in the Million Women Study. Lancet 362 (9382): 419-27, 2003. [PUBMED Abstract]
  25. Anderson GL, Limacher M, Assaf AR, et al.: Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the Women's Health Initiative randomized controlled trial. JAMA 291 (14): 1701-12, 2004. [PUBMED Abstract]
  26. Schuurman AG, van den Brandt PA, Goldbohm RA: Exogenous hormone use and the risk of postmenopausal breast cancer: results from The Netherlands Cohort Study. Cancer Causes Control 6 (5): 416-24, 1995. [PUBMED Abstract]
  27. Steinberg KK, Thacker SB, Smith SJ, et al.: A meta-analysis of the effect of estrogen replacement therapy on the risk of breast cancer. JAMA 265 (15): 1985-90, 1991. [PUBMED Abstract]
  28. Sellers TA, Mink PJ, Cerhan JR, et al.: The role of hormone replacement therapy in the risk for breast cancer and total mortality in women with a family history of breast cancer. Ann Intern Med 127 (11): 973-80, 1997. [PUBMED Abstract]
  29. Stanford JL, Weiss NS, Voigt LF, et al.: Combined estrogen and progestin hormone replacement therapy in relation to risk of breast cancer in middle-aged women. JAMA 274 (2): 137-42, 1995. [PUBMED Abstract]
  30. Colditz GA, Egan KM, Stampfer MJ: Hormone replacement therapy and risk of breast cancer: results from epidemiologic studies. Am J Obstet Gynecol 168 (5): 1473-80, 1993. [PUBMED Abstract]
  31. Gorsky RD, Koplan JP, Peterson HB, et al.: Relative risks and benefits of long-term estrogen replacement therapy: a decision analysis. Obstet Gynecol 83 (2): 161-6, 1994. [PUBMED Abstract]
  32. 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 23 (31): 7804-10, 2005. [PUBMED Abstract]
  33. Helzlsouer KJ, Harris EL, Parshad R, et al.: Familial clustering of breast cancer: possible interaction between DNA repair proficiency and radiation exposure in the development of breast cancer. Int J Cancer 64 (1): 14-7, 1995. [PUBMED Abstract]
  34. Helzlsouer KJ, Harris EL, Parshad R, et al.: DNA repair proficiency: potential susceptibility factor for breast cancer. J Natl Cancer Inst 88 (11): 754-5, 1996. [PUBMED Abstract]
  35. Abbott DW, Thompson ME, Robinson-Benion C, et al.: BRCA1 expression restores radiation resistance in BRCA1-defective cancer cells through enhancement of transcription-coupled DNA repair. J Biol Chem 274 (26): 18808-12, 1999. [PUBMED Abstract]
  36. Abbott DW, Freeman ML, Holt JT: Double-strand break repair deficiency and radiation sensitivity in BRCA2 mutant cancer cells. J Natl Cancer Inst 90 (13): 978-85, 1998. [PUBMED Abstract]
  37. Easton DF: Cancer risks in A-T heterozygotes. Int J Radiat Biol 66 (6 Suppl): S177-82, 1994. [PUBMED Abstract]
  38. Kleihues P, Schäuble B, zur Hausen A, et al.: Tumors associated with p53 germline mutations: a synopsis of 91 families. Am J Pathol 150 (1): 1-13, 1997. [PUBMED Abstract]
  39. Pierce LJ, Strawderman M, Narod SA, et al.: Effect of radiotherapy after breast-conserving treatment in women with breast cancer and germline BRCA1/2 mutations. J Clin Oncol 18 (19): 3360-9, 2000. [PUBMED Abstract]
  40. Drooger J, Akdeniz D, Pignol JP, et al.: Adjuvant radiotherapy for primary breast cancer in BRCA1 and BRCA2 mutation carriers and risk of contralateral breast cancer with special attention to patients irradiated at younger age. Breast Cancer Res Treat 154 (1): 171-80, 2015. [PUBMED Abstract]
  41. 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 7 (5): 402-6, 2006. [PUBMED Abstract]
  42. 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 24 (21): 3361-6, 2006. [PUBMED Abstract]
  43. Goldfrank D, Chuai S, Bernstein JL, et al.: Effect of mammography on breast cancer risk in women with mutations in BRCA1 or BRCA2. Cancer Epidemiol Biomarkers Prev 15 (11): 2311-3, 2006. [PUBMED Abstract]
  44. 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 112 (3): 581-4, 2008. [PUBMED Abstract]
  45. 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 345: e5660, 2012. [PUBMED Abstract]
  46. Giannakeas V, Lubinski J, Gronwald J, et al.: Mammography screening and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers: a prospective study. Breast Cancer Res Treat 147 (1): 113-8, 2014. [PUBMED Abstract]
  47. Smith-Warner SA, Spiegelman D, Yaun SS, et al.: Alcohol and breast cancer in women: a pooled analysis of cohort studies. JAMA 279 (7): 535-40, 1998. [PUBMED Abstract]
  48. Hamajima N, Hirose K, Tajima K, et al.: Alcohol, tobacco and breast cancer--collaborative reanalysis of individual data from 53 epidemiological studies, including 58,515 women with breast cancer and 95,067 women without the disease. Br J Cancer 87 (11): 1234-45, 2002. [PUBMED Abstract]
  49. McGuire V, John EM, Felberg A, et al.: No increased risk of breast cancer associated with alcohol consumption among carriers of BRCA1 and BRCA2 mutations ages <50 years. Cancer Epidemiol Biomarkers Prev 15 (8): 1565-7, 2006. [PUBMED Abstract]
  50. Dennis J, Ghadirian P, Little J, et al.: Alcohol consumption and the risk of breast cancer among BRCA1 and BRCA2 mutation carriers. Breast 19 (6): 479-83, 2010. [PUBMED Abstract]
  51. Cybulski C, Lubinski J, Huzarski T, et al.: Prospective evaluation of alcohol consumption and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 151 (2): 435-41, 2015. [PUBMED Abstract]
  52. Lammert J, Lubinski J, Gronwald J, et al.: Physical activity during adolescence and young adulthood and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 169 (3): 561-571, 2018. [PUBMED Abstract]
  53. Kehm RD, Genkinger JM, MacInnis RJ, et al.: Recreational Physical Activity Is Associated with Reduced Breast Cancer Risk in Adult Women at High Risk for Breast Cancer: A Cohort Study of Women Selected for Familial and Genetic Risk. Cancer Res 80 (1): 116-125, 2020. [PUBMED Abstract]
  54. Chen J, Pee D, Ayyagari R, et al.: Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst 98 (17): 1215-26, 2006. [PUBMED Abstract]
  55. Dupont WD, Page DL, Parl FF, et al.: Long-term risk of breast cancer in women with fibroadenoma. N Engl J Med 331 (1): 10-5, 1994. [PUBMED Abstract]
  56. Boyd NF, Byng JW, Jong RA, et al.: Quantitative classification of mammographic densities and breast cancer risk: results from the Canadian National Breast Screening Study. J Natl Cancer Inst 87 (9): 670-5, 1995. [PUBMED Abstract]
  57. Byrne C, Schairer C, Wolfe J, et al.: Mammographic features and breast cancer risk: effects with time, age, and menopause status. J Natl Cancer Inst 87 (21): 1622-9, 1995. [PUBMED Abstract]
  58. Pankow JS, Vachon CM, Kuni CC, et al.: Genetic analysis of mammographic breast density in adult women: evidence of a gene effect. J Natl Cancer Inst 89 (8): 549-56, 1997. [PUBMED Abstract]
  59. Boyd NF, Lockwood GA, Martin LJ, et al.: Mammographic densities and risk of breast cancer among subjects with a family history of this disease. J Natl Cancer Inst 91 (16): 1404-8, 1999. [PUBMED Abstract]
  60. Vachon CM, King RA, Atwood LD, et al.: Preliminary sibpair linkage analysis of percent mammographic density. J Natl Cancer Inst 91 (20): 1778-9, 1999. [PUBMED Abstract]
  61. Brunet JS, Ghadirian P, Rebbeck TR, et al.: Effect of smoking on breast cancer in carriers of mutant BRCA1 or BRCA2 genes. J Natl Cancer Inst 90 (10): 761-6, 1998. [PUBMED Abstract]
  62. Ghadirian P, Lubinski J, Lynch H, et al.: Smoking and the risk of breast cancer among carriers of BRCA mutations. Int J Cancer 110 (3): 413-6, 2004. [PUBMED Abstract]
  63. Amos CI, Struewing JP: Genetic epidemiology of epithelial ovarian cancer. Cancer 71 (2 Suppl): 566-72, 1993. [PUBMED Abstract]
  64. Stratton JF, Pharoah P, Smith SK, et al.: A systematic review and meta-analysis of family history and risk of ovarian cancer. Br J Obstet Gynaecol 105 (5): 493-9, 1998. [PUBMED Abstract]
  65. Whittemore AS, Harris R, Itnyre J: Characteristics relating to ovarian cancer risk: collaborative analysis of 12 US case-control studies. II. Invasive epithelial ovarian cancers in white women. Collaborative Ovarian Cancer Group. Am J Epidemiol 136 (10): 1184-203, 1992. [PUBMED Abstract]
  66. Brinton LA, Lamb EJ, Moghissi KS, et al.: Ovarian cancer risk after the use of ovulation-stimulating drugs. Obstet Gynecol 103 (6): 1194-203, 2004. [PUBMED Abstract]
  67. Gronwald J, Byrski T, Huzarski T, et al.: Influence of selected lifestyle factors on breast and ovarian cancer risk in BRCA1 mutation carriers from Poland. Breast Cancer Res Treat 95 (2): 105-9, 2006. [PUBMED Abstract]
  68. 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 8 (1): 26-34, 2007. [PUBMED Abstract]
  69. Antoniou AC, Rookus M, Andrieu N, et al.: Reproductive and hormonal factors, and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: results from the International BRCA1/2 Carrier Cohort Study. Cancer Epidemiol Biomarkers Prev 18 (2): 601-10, 2009. [PUBMED Abstract]
  70. Narod SA, Sun P, Ghadirian P, et al.: Tubal ligation and risk of ovarian cancer in carriers of BRCA1 or BRCA2 mutations: a case-control study. Lancet 357 (9267): 1467-70, 2001. [PUBMED Abstract]
  71. Kotsopoulos J, Lubinski J, Gronwald J, et al.: Factors influencing ovulation and the risk of ovarian cancer in BRCA1 and BRCA2 mutation carriers. Int J Cancer 137 (5): 1136-46, 2015. [PUBMED Abstract]
  72. Rodriguez C, Patel AV, Calle EE, et al.: Estrogen replacement therapy and ovarian cancer mortality in a large prospective study of US women. JAMA 285 (11): 1460-5, 2001. [PUBMED Abstract]
  73. Riman T, Dickman PW, Nilsson S, et al.: Hormone replacement therapy and the risk of invasive epithelial ovarian cancer in Swedish women. J Natl Cancer Inst 94 (7): 497-504, 2002. [PUBMED Abstract]
  74. Lacey JV, Mink PJ, Lubin JH, et al.: Menopausal hormone replacement therapy and risk of ovarian cancer. JAMA 288 (3): 334-41, 2002. [PUBMED Abstract]
  75. Anderson GL, Judd HL, Kaunitz AM, et al.: Effects of estrogen plus progestin on gynecologic cancers and associated diagnostic procedures: the Women's Health Initiative randomized trial. JAMA 290 (13): 1739-48, 2003. [PUBMED Abstract]
  76. Tortolero-Luna G, Mitchell MF: The epidemiology of ovarian cancer. J Cell Biochem Suppl 23: 200-7, 1995. [PUBMED Abstract]
  77. Hankinson SE, Hunter DJ, Colditz GA, et al.: Tubal ligation, hysterectomy, and risk of ovarian cancer. A prospective study. JAMA 270 (23): 2813-8, 1993. [PUBMED Abstract]
  78. Rutter JL, Wacholder S, Chetrit A, et al.: Gynecologic surgeries and risk of ovarian cancer in women with BRCA1 and BRCA2 Ashkenazi founder mutations: an Israeli population-based case-control study. J Natl Cancer Inst 95 (14): 1072-8, 2003. [PUBMED Abstract]
  79. Kauff ND, Satagopan JM, Robson ME, et al.: Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346 (21): 1609-15, 2002. [PUBMED Abstract]
  80. Rebbeck TR, Lynch HT, Neuhausen SL, et al.: Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 346 (21): 1616-22, 2002. [PUBMED Abstract]
  81. Kotsopoulos J, Huzarski T, Gronwald J, et al.: Bilateral Oophorectomy and Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst 109 (1): , 2017. [PUBMED Abstract]
  82. John EM, Whittemore AS, Harris R, et al.: Characteristics relating to ovarian cancer risk: collaborative analysis of seven U.S. case-control studies. Epithelial ovarian cancer in black women. Collaborative Ovarian Cancer Group. J Natl Cancer Inst 85 (2): 142-7, 1993. [PUBMED Abstract]
  83. Narod SA, Risch H, Moslehi R, et al.: Oral contraceptives and the risk of hereditary ovarian cancer. Hereditary Ovarian Cancer Clinical Study Group. N Engl J Med 339 (7): 424-8, 1998. [PUBMED Abstract]
  84. Whittemore AS, Balise RR, Pharoah PD, et al.: Oral contraceptive use and ovarian cancer risk among carriers of BRCA1 or BRCA2 mutations. Br J Cancer 91 (11): 1911-5, 2004. [PUBMED Abstract]
  85. McGuire V, Felberg A, Mills M, et al.: Relation of contraceptive and reproductive history to ovarian cancer risk in carriers and noncarriers of BRCA1 gene mutations. Am J Epidemiol 160 (7): 613-8, 2004. [PUBMED Abstract]
  86. Modan B, Hartge P, Hirsh-Yechezkel G, et al.: Parity, oral contraceptives, and the risk of ovarian cancer among carriers and noncarriers of a BRCA1 or BRCA2 mutation. N Engl J Med 345 (4): 235-40, 2001. [PUBMED Abstract]
  87. Soliman PT, Oh JC, Schmeler KM, et al.: Risk factors for young premenopausal women with endometrial cancer. Obstet Gynecol 105 (3): 575-80, 2005. [PUBMED Abstract]
  88. Vasen HF, Stormorken A, Menko FH, et al.: MSH2 mutation carriers are at higher risk of cancer than MLH1 mutation carriers: a study of hereditary nonpolyposis colorectal cancer families. J Clin Oncol 19 (20): 4074-80, 2001. [PUBMED Abstract]
  89. Daniels MS: Genetic testing by cancer site: uterus. Cancer J 18 (4): 338-42, 2012 Jul-Aug. [PUBMED Abstract]
  90. Dunlop MG, Farrington SM, Nicholl I, et al.: Population carrier frequency of hMSH2 and hMLH1 mutations. Br J Cancer 83 (12): 1643-5, 2000. [PUBMED Abstract]
  91. de la Chapelle A: The incidence of Lynch syndrome. Fam Cancer 4 (3): 233-7, 2005. [PUBMED Abstract]
  92. Ring KL, Bruegl AS, Allen BA, et al.: Germline multi-gene hereditary cancer panel testing in an unselected endometrial cancer cohort. Mod Pathol 29 (11): 1381-1389, 2016. [PUBMED Abstract]
  93. Shu CA, Pike MC, Jotwani AR, et al.: Uterine Cancer After Risk-Reducing Salpingo-oophorectomy Without Hysterectomy in Women With BRCA Mutations. JAMA Oncol 2 (11): 1434-1440, 2016. [PUBMED Abstract]
  94. 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 113 (9): 1203-1211, 2021. [PUBMED Abstract]
  95. McPherson CP, Sellers TA, Potter JD, et al.: Reproductive factors and risk of endometrial cancer. The Iowa Women's Health Study. Am J Epidemiol 143 (12): 1195-202, 1996. [PUBMED Abstract]
  96. Dossus L, Allen N, Kaaks R, et al.: Reproductive risk factors and endometrial cancer: the European Prospective Investigation into Cancer and Nutrition. Int J Cancer 127 (2): 442-51, 2010. [PUBMED Abstract]
  97. Shapiro S, Kelly JP, Rosenberg L, et al.: Risk of localized and widespread endometrial cancer in relation to recent and discontinued use of conjugated estrogens. N Engl J Med 313 (16): 969-72, 1985. [PUBMED Abstract]
  98. Ziel HK, Finkle WD: Increased risk of endometrial carcinoma among users of conjugated estrogens. N Engl J Med 293 (23): 1167-70, 1975. [PUBMED Abstract]
  99. Fisher B, Costantino JP, Redmond CK, et al.: Endometrial cancer in tamoxifen-treated breast cancer patients: findings from the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14. J Natl Cancer Inst 86 (7): 527-37, 1994. [PUBMED Abstract]
  100. Pike MC, Peters RK, Cozen W, et al.: Estrogen-progestin replacement therapy and endometrial cancer. J Natl Cancer Inst 89 (15): 1110-6, 1997. [PUBMED Abstract]
  101. Fournier A, Dossus L, Mesrine S, et al.: Risks of endometrial cancer associated with different hormone replacement therapies in the E3N cohort, 1992-2008. Am J Epidemiol 180 (5): 508-17, 2014. [PUBMED Abstract]
  102. Weiss NS, Sayvetz TA: Incidence of endometrial cancer in relation to the use of oral contraceptives. N Engl J Med 302 (10): 551-4, 1980. [PUBMED Abstract]
  103. Soini T, Hurskainen R, Grénman S, et al.: Cancer risk in women using the levonorgestrel-releasing intrauterine system in Finland. Obstet Gynecol 124 (2 Pt 1): 292-9, 2014. [PUBMED Abstract]
  104. Lindor NM, McMaster ML, Lindor CJ, et al.: Concise handbook of familial cancer susceptibility syndromes - second edition. J Natl Cancer Inst Monogr (38): 1-93, 2008. [PUBMED Abstract]
  105. Vasen HF, Offerhaus GJ, den Hartog Jager FC, et al.: The tumour spectrum in hereditary non-polyposis colorectal cancer: a study of 24 kindreds in the Netherlands. Int J Cancer 46 (1): 31-4, 1990. [PUBMED Abstract]
  106. Watson P, Lynch HT: Extracolonic cancer in hereditary nonpolyposis colorectal cancer. Cancer 71 (3): 677-85, 1993. [PUBMED Abstract]
  107. Watson P, Vasen HF, Mecklin JP, et al.: The risk of endometrial cancer in hereditary nonpolyposis colorectal cancer. Am J Med 96 (6): 516-20, 1994. [PUBMED Abstract]
  108. Aarnio M, Mecklin JP, Aaltonen LA, et al.: Life-time risk of different cancers in hereditary non-polyposis colorectal cancer (HNPCC) syndrome. Int J Cancer 64 (6): 430-3, 1995. [PUBMED Abstract]
  109. Raymond VM, Everett JN, Furtado LV, et al.: Adrenocortical carcinoma is a lynch syndrome-associated cancer. J Clin Oncol 31 (24): 3012-8, 2013. [PUBMED Abstract]
  110. Raymond VM, Mukherjee B, Wang F, et al.: Elevated risk of prostate cancer among men with Lynch syndrome. J Clin Oncol 31 (14): 1713-8, 2013. [PUBMED Abstract]
  111. Suspiro A, Fidalgo P, Cravo M, et al.: The Muir-Torre syndrome: a rare variant of hereditary nonpolyposis colorectal cancer associated with hMSH2 mutation. Am J Gastroenterol 93 (9): 1572-4, 1998. [PUBMED Abstract]
  112. Kerber RA, Slattery ML: Comparison of self-reported and database-linked family history of cancer data in a case-control study. Am J Epidemiol 146 (3): 244-8, 1997. [PUBMED Abstract]
  113. Parent ME, Ghadirian P, Lacroix A, et al.: The reliability of recollections of family history: implications for the medical provider. J Cancer Educ 12 (2): 114-20, 1997 Summer. [PUBMED Abstract]
  114. Ready K, Litton JK, Arun BK: Clinical application of breast cancer risk assessment models. Future Oncol 6 (3): 355-65, 2010. [PUBMED Abstract]
  115. Amir E, Freedman OC, Seruga B, et al.: Assessing women at high risk of breast cancer: a review of risk assessment models. J Natl Cancer Inst 102 (10): 680-91, 2010. [PUBMED Abstract]
  116. Rosner BA, Colditz GA, Webb PM, et al.: Mathematical models of ovarian cancer incidence. Epidemiology 16 (4): 508-15, 2005. [PUBMED Abstract]
  117. Pfeiffer RM, Park Y, Kreimer AR, et al.: Risk prediction for breast, endometrial, and ovarian cancer in white women aged 50 y or older: derivation and validation from population-based cohort studies. PLoS Med 10 (7): e1001492, 2013. [PUBMED Abstract]
  118. Gail MH, Mai PL: Comparing breast cancer risk assessment models. J Natl Cancer Inst 102 (10): 665-8, 2010. [PUBMED Abstract]
  119. Quante AS, Whittemore AS, Shriver T, et al.: Breast cancer risk assessment across the risk continuum: genetic and nongenetic risk factors contributing to differential model performance. Breast Cancer Res 14 (6): R144, 2012. [PUBMED Abstract]
  120. Gail MH, Brinton LA, Byar DP, et al.: Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. J Natl Cancer Inst 81 (24): 1879-86, 1989. [PUBMED Abstract]
  121. Colditz GA, Rosner B: Cumulative risk of breast cancer to age 70 years according to risk factor status: data from the Nurses' Health Study. Am J Epidemiol 152 (10): 950-64, 2000. [PUBMED Abstract]
  122. Claus EB, Risch N, Thompson WD: Autosomal dominant inheritance of early-onset breast cancer. Implications for risk prediction. Cancer 73 (3): 643-51, 1994. [PUBMED Abstract]
  123. Tyrer J, Duffy SW, Cuzick J: A breast cancer prediction model incorporating familial and personal risk factors. Stat Med 23 (7): 1111-30, 2004. [PUBMED Abstract]
  124. Parmigiani G, Berry D, Aguilar O: Determining carrier probabilities for breast cancer-susceptibility genes BRCA1 and BRCA2. Am J Hum Genet 62 (1): 145-58, 1998. [PUBMED Abstract]
  125. Antoniou AC, Pharoah PD, McMullan G, et al.: A comprehensive model for familial breast cancer incorporating BRCA1, BRCA2 and other genes. Br J Cancer 86 (1): 76-83, 2002. [PUBMED Abstract]
  126. Antoniou AC, Pharoah PP, Smith P, et al.: The BOADICEA model of genetic susceptibility to breast and ovarian cancer. Br J Cancer 91 (8): 1580-90, 2004. [PUBMED Abstract]
  127. Antoniou AC, Cunningham AP, Peto J, et al.: The BOADICEA model of genetic susceptibility to breast and ovarian cancers: updates and extensions. Br J Cancer 98 (8): 1457-66, 2008. [PUBMED Abstract]
  128. Anothaisintawee T, Teerawattananon Y, Wiratkapun C, et al.: Risk prediction models of breast cancer: a systematic review of model performances. Breast Cancer Res Treat 133 (1): 1-10, 2012. [PUBMED Abstract]
  129. Amir E, Evans DG, Shenton A, et al.: Evaluation of breast cancer risk assessment packages in the family history evaluation and screening programme. J Med Genet 40 (11): 807-14, 2003. [PUBMED Abstract]
  130. Laitman Y, Simeonov M, Keinan-Boker L, et al.: Breast cancer risk prediction accuracy in Jewish Israeli high-risk women using the BOADICEA and IBIS risk models. Genet Res (Camb) 95 (6): 174-7, 2013. [PUBMED Abstract]
  131. MacInnis RJ, Bickerstaffe A, Apicella C, et al.: Prospective validation of the breast cancer risk prediction model BOADICEA and a batch-mode version BOADICEACentre. Br J Cancer 109 (5): 1296-301, 2013. [PUBMED Abstract]
  132. Terry MB, Liao Y, Whittemore AS, et al.: 10-year performance of four models of breast cancer risk: a validation study. Lancet Oncol 20 (4): 504-517, 2019. [PUBMED Abstract]
  133. Claus EB, Risch N, Thompson WD: The calculation of breast cancer risk for women with a first degree family history of ovarian cancer. Breast Cancer Res Treat 28 (2): 115-20, 1993. [PUBMED Abstract]
  134. Bondy ML, Lustbader ED, Halabi S, et al.: Validation of a breast cancer risk assessment model in women with a positive family history. J Natl Cancer Inst 86 (8): 620-5, 1994. [PUBMED Abstract]
  135. Spiegelman D, Colditz GA, Hunter D, et al.: Validation of the Gail et al. model for predicting individual breast cancer risk. J Natl Cancer Inst 86 (8): 600-7, 1994. [PUBMED Abstract]
  136. Rockhill B, Spiegelman D, Byrne C, et al.: Validation of the Gail et al. model of breast cancer risk prediction and implications for chemoprevention. J Natl Cancer Inst 93 (5): 358-66, 2001. [PUBMED Abstract]
  137. Costantino JP, Gail MH, Pee D, et al.: Validation studies for models projecting the risk of invasive and total breast cancer incidence. J Natl Cancer Inst 91 (18): 1541-8, 1999. [PUBMED Abstract]
  138. Bondy ML, Newman LA: Breast cancer risk assessment models: applicability to African-American women. Cancer 97 (1 Suppl): 230-5, 2003. [PUBMED Abstract]
  139. Schonfeld SJ, Pee D, Greenlee RT, et al.: Effect of changing breast cancer incidence rates on the calibration of the Gail model. J Clin Oncol 28 (14): 2411-7, 2010. [PUBMED Abstract]
  140. Gail MH, Costantino JP, Pee D, et al.: Projecting individualized absolute invasive breast cancer risk in African American women. J Natl Cancer Inst 99 (23): 1782-92, 2007. [PUBMED Abstract]
  141. Gail MH: Discriminatory accuracy from single-nucleotide polymorphisms in models to predict breast cancer risk. J Natl Cancer Inst 100 (14): 1037-41, 2008. [PUBMED Abstract]
  142. Gail MH: Value of adding single-nucleotide polymorphism genotypes to a breast cancer risk model. J Natl Cancer Inst 101 (13): 959-63, 2009. [PUBMED Abstract]
  143. Barlow WE, White E, Ballard-Barbash R, et al.: Prospective breast cancer risk prediction model for women undergoing screening mammography. J Natl Cancer Inst 98 (17): 1204-14, 2006. [PUBMED Abstract]
  144. Tice JA, Cummings SR, Ziv E, et al.: Mammographic breast density and the Gail model for breast cancer risk prediction in a screening population. Breast Cancer Res Treat 94 (2): 115-22, 2005. [PUBMED Abstract]
  145. Lee AJ, Cunningham AP, Tischkowitz M, et al.: Incorporating truncating variants in PALB2, CHEK2, and ATM into the BOADICEA breast cancer risk model. Genet Med 18 (12): 1190-1198, 2016. [PUBMED Abstract]
  146. MacInnis RJ, Liao Y, Knight JA, et al.: Considerations When Using Breast Cancer Risk Models for Women with Negative BRCA1/BRCA2 Mutation Results. J Natl Cancer Inst 112 (4): 418-422, 2020. [PUBMED Abstract]
  147. Barnetson RA, Tenesa A, Farrington SM, et al.: Identification and survival of carriers of mutations in DNA mismatch-repair genes in colon cancer. N Engl J Med 354 (26): 2751-63, 2006. [PUBMED Abstract]
  148. Kastrinos F, Uno H, Ukaegbu C, et al.: Development and Validation of the PREMM5 Model for Comprehensive Risk Assessment of Lynch Syndrome. J Clin Oncol 35 (19): 2165-2172, 2017. [PUBMED Abstract]
  149. Chen S, Wang W, Lee S, et al.: Prediction of germline mutations and cancer risk in the Lynch syndrome. JAMA 296 (12): 1479-87, 2006. [PUBMED Abstract]
  150. Khan O, Blanco A, Conrad P, et al.: Performance of Lynch syndrome predictive models in a multi-center US referral population. Am J Gastroenterol 106 (10): 1822-7; quiz 1828, 2011. [PUBMED Abstract]
  151. Mercado RC, Hampel H, Kastrinos F, et al.: Performance of PREMM(1,2,6), MMRpredict, and MMRpro in detecting Lynch syndrome among endometrial cancer cases. Genet Med 14 (7): 670-80, 2012. [PUBMED Abstract]
  152. Robson ME, Storm CD, Weitzel J, et al.: American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 28 (5): 893-901, 2010. [PUBMED Abstract]
  153. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2022. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2022. Available online with free registration. Last accessed March 16, 2022.
  154. Statement of the American Society of Human Genetics on genetic testing for breast and ovarian cancer predisposition. Am J Hum Genet 55 (5): i-iv, 1994. [PUBMED Abstract]
  155. Hampel H, Bennett RL, Buchanan A, et al.: A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med 17 (1): 70-87, 2015. [PUBMED Abstract]
  156. Owens DK, Davidson KW, Krist AH, et al.: Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 322 (7): 652-665, 2019. [PUBMED Abstract]
  157. Lancaster JM, Powell CB, Kauff ND, et al.: Society of Gynecologic Oncologists Education Committee statement on risk assessment for inherited gynecologic cancer predispositions. Gynecol Oncol 107 (2): 159-62, 2007. [PUBMED Abstract]
  158. Manahan ER, Kuerer HM, Sebastian M, et al.: Consensus Guidelines on Genetic` Testing for Hereditary Breast Cancer from the American Society of Breast Surgeons. Ann Surg Oncol 26 (10): 3025-3031, 2019. [PUBMED Abstract]
  159. Beitsch PD, Whitworth PW, Hughes K, et al.: Underdiagnosis of Hereditary Breast Cancer: Are Genetic Testing Guidelines a Tool or an Obstacle? J Clin Oncol 37 (6): 453-460, 2019. [PUBMED Abstract]
  160. Cropper C, Woodson A, Arun B, et al.: Evaluating the NCCN Clinical Criteria for Recommending BRCA1 and BRCA2 Genetic Testing in Patients With Breast Cancer. J Natl Compr Canc Netw 15 (6): 797-803, 2017. [PUBMED Abstract]
  161. Grindedal EM, Heramb C, Karsrud I, et al.: Current guidelines for BRCA testing of breast cancer patients are insufficient to detect all mutation carriers. BMC Cancer 17 (1): 438, 2017. [PUBMED Abstract]
  162. Yadav S, Hu C, Hart SN, et al.: Evaluation of Germline Genetic Testing Criteria in a Hospital-Based Series of Women With Breast Cancer. J Clin Oncol 38 (13): 1409-1418, 2020. [PUBMED Abstract]
  163. Kurian AW, Bernhisel R, Larson K, et al.: Prevalence of Pathogenic Variants in Cancer Susceptibility Genes Among Women With Postmenopausal Breast Cancer. JAMA 323 (10): 995-997, 2020. [PUBMED Abstract]
  164. Couch FJ, Nathanson KL, Offit K: Two decades after BRCA: setting paradigms in personalized cancer care and prevention. Science 343 (6178): 1466-70, 2014. [PUBMED Abstract]
  165. Theobald KA, Susswein LR, Marshall ML, et al.: Utility of Expedited Hereditary Cancer Testing in the Surgical Management of Patients with a New Breast Cancer Diagnosis. Ann Surg Oncol 25 (12): 3556-3562, 2018. [PUBMED Abstract]
  166. Gornick MC, Kurian AW, An LC, et al.: Knowledge regarding and patterns of genetic testing in patients newly diagnosed with breast cancer participating in the iCanDecide trial. Cancer 124 (20): 4000-4009, 2018. [PUBMED Abstract]
  167. Chiba A, Hoskin TL, Hallberg EJ, et al.: Impact that Timing of Genetic Mutation Diagnosis has on Surgical Decision Making and Outcome for BRCA1/BRCA2 Mutation Carriers with Breast Cancer. Ann Surg Oncol 23 (10): 3232-8, 2016. [PUBMED Abstract]
  168. Obermair A, Youlden DR, Baade PD, et al.: The impact of risk-reducing hysterectomy and bilateral salpingo-oophorectomy on survival in patients with a history of breast cancer--a population-based data linkage study. Int J Cancer 134 (9): 2211-22, 2014. [PUBMED Abstract]
  169. Byrski T, Dent R, Blecharz P, et al.: Results of a phase II open-label, non-randomized trial of cisplatin chemotherapy in patients with BRCA1-positive metastatic breast cancer. Breast Cancer Res 14 (4): R110, 2012. [PUBMED Abstract]
  170. Robson M, Im SA, Senkus E, et al.: Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 377 (6): 523-533, 2017. [PUBMED Abstract]
  171. Heymann S, Delaloge S, Rahal A, et al.: Radio-induced malignancies after breast cancer postoperative radiotherapy in patients with Li-Fraumeni syndrome. Radiat Oncol 5: 104, 2010. [PUBMED Abstract]
  172. Bougeard G, Renaux-Petel M, Flaman JM, et al.: Revisiting Li-Fraumeni Syndrome From TP53 Mutation Carriers. J Clin Oncol 33 (21): 2345-52, 2015. [PUBMED Abstract]
  173. Rebbeck TR, Kauff ND, Domchek SM: Meta-analysis of risk reduction estimates associated with risk-reducing salpingo-oophorectomy in BRCA1 or BRCA2 mutation carriers. J Natl Cancer Inst 101 (2): 80-7, 2009. [PUBMED Abstract]
  174. Kotsopoulos J, Lubinski J, Lynch HT, et al.: Oophorectomy after menopause and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev 21 (7): 1089-96, 2012. [PUBMED Abstract]
  175. Cass I, Baldwin RL, Varkey T, et al.: Improved survival in women with BRCA-associated ovarian carcinoma. Cancer 97 (9): 2187-95, 2003. [PUBMED Abstract]
  176. Tan DS, Rothermundt C, Thomas K, et al.: "BRCAness" syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol 26 (34): 5530-6, 2008. [PUBMED Abstract]
  177. Moore K, Colombo N, Scambia G, et al.: Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 379 (26): 2495-2505, 2018. [PUBMED Abstract]
  178. Practice Bulletin No. 149: Endometrial cancer. Obstet Gynecol 125 (4): 1006-26, 2015. [PUBMED Abstract]
  179. Ott PA, Bang YJ, Berton-Rigaud D, et al.: Safety and Antitumor Activity of Pembrolizumab in Advanced Programmed Death Ligand 1-Positive Endometrial Cancer: Results From the KEYNOTE-028 Study. J Clin Oncol 35 (22): 2535-2541, 2017. [PUBMED Abstract]
  180. Walsh T, Casadei S, Lee MK, et al.: Mutations in 12 genes for inherited ovarian, fallopian tube, and peritoneal carcinoma identified by massively parallel sequencing. Proc Natl Acad Sci U S A 108 (44): 18032-7, 2011. [PUBMED Abstract]
  181. Frey MK, Kim SH, Bassett RY, et al.: Rescreening for genetic mutations using multi-gene panel testing in patients who previously underwent non-informative genetic screening. Gynecol Oncol 139 (2): 211-5, 2015. [PUBMED Abstract]
  182. Desmond A, Kurian AW, Gabree M, et al.: Clinical Actionability of Multigene Panel Testing for Hereditary Breast and Ovarian Cancer Risk Assessment. JAMA Oncol 1 (7): 943-51, 2015. [PUBMED Abstract]
  183. Couch FJ, Shimelis H, Hu C, et al.: Associations Between Cancer Predisposition Testing Panel Genes and Breast Cancer. JAMA Oncol 3 (9): 1190-1196, 2017. [PUBMED Abstract]
  184. Weitzel JN, Neuhausen SL, Adamson A, et al.: Pathogenic and likely pathogenic variants in PALB2, CHEK2, and other known breast cancer susceptibility genes among 1054 BRCA-negative Hispanics with breast cancer. Cancer 125 (16): 2829-2836, 2019. [PUBMED Abstract]
  185. Frey MK, Kopparam RV, Ni Zhou Z, et al.: Prevalence of nonfounder BRCA1/2 mutations in Ashkenazi Jewish patients presenting for genetic testing at a hereditary breast and ovarian cancer center. Cancer 125 (5): 690-697, 2019. [PUBMED Abstract]
  186. Eoh KJ, Kim JE, Park HS, et al.: Detection of Germline Mutations in Patients with Epithelial Ovarian Cancer Using Multi-gene Panels: Beyond BRCA1/2. Cancer Res Treat 50 (3): 917-925, 2018. [PUBMED Abstract]
  187. Tung N, Lin NU, Kidd J, et al.: Frequency of Germline Mutations in 25 Cancer Susceptibility Genes in a Sequential Series of Patients With Breast Cancer. J Clin Oncol 34 (13): 1460-8, 2016. [PUBMED Abstract]
  188. Buys SS, Sandbach JF, Gammon A, et al.: A study of over 35,000 women with breast cancer tested with a 25-gene panel of hereditary cancer genes. Cancer 123 (10): 1721-1730, 2017. [PUBMED Abstract]
  189. Pritzlaff M, Summerour P, McFarland R, et al.: Male breast cancer in a multi-gene panel testing cohort: insights and unexpected results. Breast Cancer Res Treat 161 (3): 575-586, 2017. [PUBMED Abstract]
  190. Yadav S, Reeves A, Campian S, et al.: Outcomes of retesting BRCA negative patients using multigene panels. Fam Cancer 16 (3): 319-328, 2017. [PUBMED Abstract]
  191. Crawford B, Adams SB, Sittler T, et al.: Multi-gene panel testing for hereditary cancer predisposition in unsolved high-risk breast and ovarian cancer patients. Breast Cancer Res Treat 163 (2): 383-390, 2017. [PUBMED Abstract]
  192. Kurian AW, Ward KC, Howlader N, et al.: Genetic Testing and Results in a Population-Based Cohort of Breast Cancer Patients and Ovarian Cancer Patients. J Clin Oncol 37 (15): 1305-1315, 2019. [PUBMED Abstract]
  193. Carter NJ, Marshall ML, Susswein LR, et al.: Germline pathogenic variants identified in women with ovarian tumors. Gynecol Oncol 151 (3): 481-488, 2018. [PUBMED Abstract]
  194. Shimelis H, LaDuca H, Hu C, et al.: Triple-Negative Breast Cancer Risk Genes Identified by Multigene Hereditary Cancer Panel Testing. J Natl Cancer Inst 110 (8): 855-862, 2018. [PUBMED Abstract]
  195. Southey MC, Goldgar DE, Winqvist R, et al.: PALB2, CHEK2 and ATM rare variants and cancer risk: data from COGS. J Med Genet 53 (12): 800-811, 2016. [PUBMED Abstract]
  196. Kurian AW, Ward KC, Hamilton AS, et al.: Uptake, Results, and Outcomes of Germline Multiple-Gene Sequencing After Diagnosis of Breast Cancer. JAMA Oncol 4 (8): 1066-1072, 2018. [PUBMED Abstract]

Penetrance of Inherited Susceptibility to Hereditary Breast and/or Gynecologic Cancers

The proportion of individuals carrying a pathogenic variant who will manifest a certain disease is referred to as penetrance. In general, common genetic variants that are associated with cancer susceptibility have a lower penetrance than rare genetic variants. This is depicted in Figure 4. For adult-onset diseases, penetrance is usually described by the individual carrier's age, sex, and organ site. For example, the penetrance for breast cancer in female carriers of BRCA1 pathogenic variants is often quoted by age 50 years and by age 70 years. Of the numerous methods for estimating penetrance, none are without potential biases, and determining an individual carrier's risk of cancer involves some level of imprecision.

Enlarge Graph shows relative risk on the x-axis and allele frequency on the y-axis. A line depicts the general finding of a low relative risk associated with common, low-penetrance genetic variants and a higher relative risk associated with rare, high-penetrance genetic variants.
Figure 4. Genetic architecture of cancer risk. This graph depicts the general finding of a low relative risk associated with common, low-penetrance genetic variants, such as single-nucleotide polymorphisms identified in genome-wide association studies, and a higher relative risk associated with rare, high-penetrance genetic variants, such as pathogenic variants in the BRCA1/BRCA2 genes associated with hereditary breast and ovarian cancer and the mismatch repair genes associated with Lynch syndrome.

Throughout this summary, we discuss studies that report on relative and absolute risks. These are two important but different concepts. Relative risk (RR) refers to an estimate of risk relative to another group (e.g., risk of an outcome like breast cancer for women who are exposed to a risk factor relative to the risk of breast cancer for women who are unexposed to the same risk factor). RR measures that are greater than 1 mean that the risk for those captured in the numerator (i.e., the exposed) is higher than the risk for those captured in the denominator (i.e., the unexposed). RR measures that are less than 1 mean that the risk for those captured in the numerator (i.e., the exposed) is lower than the risk for those captured in the denominator (i.e., the unexposed). Measures with similar relative interpretations include the odds ratio (OR), hazard ratio, and risk ratio.

Absolute risk measures consider the number of people who have a particular outcome, the number of people in a population who could have the outcome, and person-time (the period of time during which an individual was at risk of having the outcome). Absolute risk measures also reflect the absolute burden of an outcome in a population. Absolute measures include risks and rates and can be expressed over a specific time frame (e.g., 1 year, 5 years) or overall lifetime. Cumulative risk is a measure of risk that occurs over a defined time period. For example, overall lifetime risk is a type of cumulative risk that is usually calculated on the basis of a given life expectancy (e.g., 80 or 90 years). Cumulative risk can also be presented over other time frames (e.g., up to age 50 years).

Large relative risk measures do not mean that there will be large effects in the actual number of individuals at a population level because the disease outcome may be quite rare. For example, the relative risk for smoking is much higher for lung cancer than for heart disease, but the absolute difference between smokers and nonsmokers is greater for heart disease, the more-common outcome, than for lung cancer, the more-rare outcome.

Therefore, in evaluating the effect of exposures and biological markers on disease prevention across the continuum, it is important to recognize the differences between relative and absolute effects in weighing the overall impact of a given risk factor. For example, the magnitude is in the range of 30% (e.g., ORs or RRs of 1.3) for many breast cancer risk factors, which means that women with a risk factor (e.g., alcohol consumption, late age at first birth, oral contraceptive use, postmenopausal body size) have a 30% relative increase in breast cancer in comparison with what they would have if they did not have that risk factor. But the absolute increase in risk is based on the underlying absolute risk of disease. Figure 5 and Table 2 show the impact of a relative risk factor in the range of 1.3 on absolute risk. (Refer to the Standard Pedigree Nomenclature figure in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for definitions of the standard symbols used in these pedigrees.) As shown, women with a family history of breast cancer have a much higher benefit from risk factor reduction on an absolute scale.[1]

Enlarge Five pedigrees are shown depicting probands with varying degrees of family history of breast cancer ranging from no affected first-degree relatives and no known BRCA mutation in the family (family 1) to three affected first-degree relatives, including one relative with bilateral breast cancer, and a known BRCA1 mutation in the family (family 5).
Figure 5. These five pedigrees depict probands with varying degrees of family history. Table 2 accompanies this figure.
Table 2. Effect of Altering a Risk Factor With Relative Risk of 1.3 Across Women With Different Family Histories of Breast Cancer a
Family History Lifetime Risk (%) Lifetime Risk After Risk Factor Modification (%) Absolute Risk Difference (%) Relative Risk
aRefer to Figure 5, which accompanies this table.
Low (Family 1) 10.9  8.4 2.50 1.29 (29% increased risk)
Moderate (Family 2) 21.6 16.8 4.80 1.28 (28% increased risk)
Moderate/high (Family 3) 27.1 21.3 5.80 1.27 (27% increased risk)
High (Family 4) 32.0 25.3 6.70 1.26 (26% increased risk)
BRCA1 pathogenic variant (Family 5) 53.7 44.2 9.50 1.21 (21% increased risk)

With the increasing use of multigene panel tests, a framework for cancer risk management among individuals with pathogenic variants detected in novel genes has been described [2] that incorporates data on age-specific, lifetime, and absolute cancer risks. The framework suggests initiating screening in these individuals at the age when their 5-year cancer risk approaches that at which screening is routinely initiated for women in the general population (approximately 1% for breast cancer in the United States). As a result, the age at which to begin screening will vary depending on the gene. (Refer to the Multigene [panel] testing section in the Introduction section of this summary for more information on multigene panel tests.)

References
  1. Quante AS, Herz J, Whittemore AS, et al.: Assessing absolute changes in breast cancer risk due to modifiable risk factors. Breast Cancer Res Treat 152 (1): 193-7, 2015. [PUBMED Abstract]
  2. Tung N, Domchek SM, Stadler Z, et al.: Counselling framework for moderate-penetrance cancer-susceptibility mutations. Nat Rev Clin Oncol 13 (9): 581-8, 2016. [PUBMED Abstract]

Genes Associated With Breast and/or Gynecologic Cancer Susceptibility

Several genes are found to be associated with the development of breast and/or gynecologic cancers. These genes are categorized as high-penetrance, moderate-penetrance, and low-penetrance in this summary. The high- and moderate-penetrance genes are summarized in Table 3. Low-penetrance genes and loci primarily include polymorphisms that have been associated with cancer susceptibility. (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes, Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer, and Single Nucleotide Variant–Associated Cancer Risks sections of this summary for more information.)

Table 3. Genes Associated With Breast and/or Gynecologic Cancer Susceptibility
Cancer Susceptibilitya Moderate-Penetrance Genesb High-Penetrance Genes
aOther cancers may be associated with the genes in this table.
bOther genes discussed in the Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer section of this summary but for which penetrance is unknown include CASP8, TGFB1, Abraxas, RECQL, and SMARCA4.
Breast cancer ATM, BRIP1, CHEK2, FANCD2, RAD51C BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11, TP53
Ovarian cancer ATM, BRIP1, EPCAM, MLH1, MSH2, MSH6, RAD51C BRCA1, BRCA2
Endometrial cancer   EPCAM, MLH1, MSH2, MSH6, PMS2, PTEN

High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes

BRCA1 and BRCA2

Introduction

Epidemiologic studies have clearly established the role of family history as an important risk factor for both breast and ovarian cancers. After gender and age, a positive family history is the strongest known predictive risk factor for breast cancer. However, it has long been recognized that in some families, there is hereditary breast cancer, which is characterized by an early age of onset, bilaterality, and the presence of breast cancer in multiple generations in an apparent autosomal dominant pattern of transmission (through either the maternal or the paternal lineage), sometimes including tumors of other organs, particularly the ovary and prostate gland.[1,2] It is now known that the cancer in some of these families can be explained by specific pathogenic variants in single cancer susceptibility genes. The isolation of several of these genes, which when altered are associated with a significantly increased risk of breast/ovarian cancer, makes it possible to identify individuals at risk. Although such cancer susceptibility genes are very important, highly penetrant  germline pathogenic variants are estimated to account for only 5% to 10% of breast cancers overall.

A 1988 study reported the first quantitative evidence that breast cancer segregated as an autosomal dominant trait in some families.[3] The search for genes associated with hereditary susceptibility to breast cancer has been facilitated by studies of large kindreds with multiple affected individuals and has led to the identification of several susceptibility genes, including BRCA1, BRCA2, TP53, PTEN/MMAC1, and STK11. Other genes, such as the mismatch repair genes MLH1, MSH2, MSH6, and PMS2, have been associated with an increased risk of ovarian cancer, but have not been consistently associated with breast cancer.

BRCA1

In 1990, a susceptibility gene for breast cancer was mapped by genetic linkage to the long arm of chromosome 17, in the interval 17q12-21.[4] The linkage between breast cancer and genetic markers on chromosome 17q was soon confirmed by others, and evidence for the coincident transmission of both breast and ovarian cancer susceptibility in linked families was observed.[5] The BRCA1 gene was subsequently identified by positional cloning methods and has been found to contain 24 exons that encode a protein of 1,863 amino acids. Germline pathogenic variants in BRCA1 are associated with early-onset breast cancer, ovarian cancer, and fallopian tube cancer. (Refer to the Penetrance of BRCA pathogenic variants section of this summary for more information.) Male breast cancer, pancreatic cancer, testicular cancer, and early-onset prostate cancer may also be associated with pathogenic variants in BRCA1;[6-9] however, male breast cancer, pancreatic cancer, and prostate cancer are more strongly associated with pathogenic variants in BRCA2.

BRCA2

A second breast cancer susceptibility gene, BRCA2, was localized to the long arm of chromosome 13 through linkage studies of 15 families with multiple cases of breast cancer that were not linked to BRCA1. Pathogenic variants in BRCA2 are associated with multiple cases of breast cancer in families, and are also associated with male breast cancer, ovarian cancer, prostate cancer, melanoma, and pancreatic cancer.[8-14] (Refer to the Penetrance of BRCA pathogenic variants section of this summary for more information.) BRCA2 is a large gene with 27 exons that encode a protein of 3,418 amino acids.[15] While not homologous genes, both BRCA1 and BRCA2 have an unusually large exon 11 and translational start sites in exon 2. Like BRCA1, BRCA2 appears to behave like a tumor suppressor gene. In tumors associated with both BRCA1 and BRCA2 pathogenic variants, there is often loss of the wild-type allele.

Pathogenic variants in BRCA1 and BRCA2 appear to be responsible for disease in 45% of families with multiple cases of breast cancer and in up to 90% of families with both breast and ovarian cancer.[16]

BRCA1 and BRCA2 function

Most BRCA1 and BRCA2 pathogenic variants are predicted to produce a truncated protein product, and thus loss of protein function, although some missense pathogenic variants cause loss of function without truncation. Because inherited breast/ovarian cancer is an autosomal dominant condition, individuals with a BRCA1 or BRCA2 pathogenic variant on one copy of chromosome 17 or 13 also carry a normal allele on the other paired chromosome. In most breast and ovarian cancers that have been studied from carriers of pathogenic variants, deletion of the normal allele results in loss of all function, leading to the classification of BRCA1 and BRCA2 as tumor suppressor genes. In addition to their roles as tumor suppressor genes, BRCA1 and BRCA2 are involved in myriad functions within cells, including homologous DNA repair, genomic stability, transcriptional regulation, protein ubiquitination, chromatin remodeling, and cell cycle control.[17,18]

Pathogenic variants in BRCA1 and BRCA2

Nearly 2,000 distinct variants and sequence variations in BRCA1 and BRCA2 have already been described.[19] Approximately 1 in 400 to 800 individuals in the general population may carry a germline pathogenic variant in BRCA1 or BRCA2.[20,21] The variants that have been associated with increased cancer risk result in missing or nonfunctional proteins, supporting the hypothesis that BRCA1 and BRCA2 are tumor suppressor genes. While a small number of these pathogenic variants have been found repeatedly in unrelated families, most have not been reported in more than a few families.

Variant-screening methods vary in their sensitivity. Methods widely used in research laboratories, such as single-stranded conformational polymorphism analysis and conformation-sensitive gel electrophoresis, miss nearly a third of the variants that are detected by DNA sequencing.[22] In addition, large genomic alterations such as translocations, inversions, deletions, or insertions are missed by most of the techniques, including direct DNA sequencing; however, testing for these alterations is commercially available. Such rearrangements are believed to be responsible for 12% to 18% of BRCA1 inactivating variants but are seen less frequently in the BRCA2 gene and in individuals of Ashkenazi Jewish (AJ) descent.[23-29] Furthermore, studies have suggested that these rearrangements may be more frequently seen in Hispanic and Caribbean populations.[27,29,30]

Variants of uncertain significance

Germline pathogenic variants in the BRCA1/BRCA2 genes are associated with an approximately 60% lifetime risk of breast cancer and a 15% to 40% lifetime risk of ovarian cancer. There are no definitive functional tests for BRCA1 or BRCA2; therefore, the classification of nucleotide changes to predict their functional impact as deleterious or benign relies on imperfect data. The majority of accepted pathogenic variants result in protein truncation and/or loss of important functional domains. However, 10% to 15% of all individuals undergoing genetic testing with full sequencing of BRCA1 and BRCA2 will not have a clearly pathogenic variant detected; instead, they will have a variant of uncertain (or unknown) significance (VUS). VUS may cause substantial challenges in genetic counseling, particularly in terms of cancer risk estimates and risk management. Clinical management of such patients needs to be highly individualized and must take into consideration factors such as the patient’s personal and family cancer history, in addition to sources of information to help characterize the VUS as benign or deleterious. Thus an improved classification and reporting system may be of clinical utility.[31]

A comprehensive analysis of 7,461 consecutive full gene sequence analyses performed by Myriad Genetic Laboratories, Inc., described the frequency of VUS found over a 3-year period.[32] Among subjects who did not have a clearly pathogenic variant, 13% had VUS defined as “missense mutations and mutations that occur in analyzed intronic regions whose clinical significance has not yet been determined, chain-terminating mutations that truncate BRCA1 and BRCA2 distal to amino acid positions 1853 and 3308, respectively, and mutations that eliminate the normal stop codons for these proteins.” The classification of a sequence variant as a VUS is a moving target. An additional 6.8% of subjects without clear pathogenic variants had sequence alterations that were once considered VUS but were reclassified as a polymorphisms, or occasionally as pathogenic variants.

The frequency of VUS varies by ethnicity within the U.S. population. African American individuals appear to have the highest rate of VUS.[33] In a 2009 study of data from Myriad, 16.5% of individuals of African ancestry had VUS, the highest rate among all ethnicities. The frequency of VUS in Asian, Middle Eastern, and Hispanic populations clusters between 10% and 14%, although these numbers are based on limited sample sizes. Over time, the rate of changes classified as VUS has decreased in all ethnicities, largely the result of improved variant classification algorithms.[34] VUS continue to be reclassified as additional information is curated and interpreted.[35,36] Such information may impact the continuing care of affected individuals.

A number of methods for discriminating deleterious from neutral VUS exist and others are in development [37-40] including integrated methods (see below).[41] Interpretation of VUS is greatly aided by efforts to determine if there is cosegregation of a VUS with the cancers seen in a family. In general, a VUS observed in individuals who also have a pathogenic variant, especially when the same VUS has been identified in conjunction with different pathogenic variants, is less likely to be deleterious, although there are rare exceptions. As an adjunct to the clinical information, models have been deployed to interpret VUS. These models analyze VUS based on sequence conservation, biochemical properties of amino acid changes,[37,42-46] incorporation of information on pathologic characteristics of BRCA1- and BRCA2-related tumors (e.g., BRCA1-related breast cancers are usually estrogen receptor [ER]–negative),[47] and functional studies to measure the influence of specific sequence variations on the activity of BRCA1 or BRCA2 proteins.[48,49] When attempting to interpret a VUS, all available information should be examined.

Population estimates of the likelihood of having a BRCA1 or BRCA2 pathogenic variant

Statistics regarding the percentage of individuals found to be carriers of BRCA pathogenic variants among samples of women and men with a variety of personal cancer histories regardless of family history are provided below. These data can help determine who might best benefit from a referral for cancer genetic counseling and consideration of genetic testing but cannot replace a personalized risk assessment, which might indicate a higher or lower pathogenic variant likelihood based on additional personal and family history characteristics.

In some cases, the same pathogenic variant has been found in multiple unrelated families. This observation is consistent with a founder effect, wherein a pathogenic variant identified in a contemporary population can be traced to a small group of founders isolated by geographic, cultural, or other factors. Most notably, two specific BRCA1 pathogenic variants (185delAG and 5382insC) and a BRCA2 pathogenic variant (6174delT) have been reported to be common in AJs. However, other founder pathogenic variants have been identified in African American and Hispanic individuals.[30,50,51] The presence of these founder pathogenic variants has practical implications for genetic testing. Many laboratories offer directed testing specifically for ethnic-specific alleles. This greatly simplifies the technical aspects of the test but is not without limitations. Nonfounder BRCA pathogenic variants in the AJ population have been reported to be between 3% and 15%.[32,52,53]

Among the general population, the likelihood of having any BRCA pathogenic variant is as follows:

  • General population (excluding Ashkenazim): about 1 in 400 (~0.25%).[21,54]
  • Women with breast cancer (any age): 1 in 50 (2%).[55]
  • Women with breast cancer (younger than 40 y): 1 in 10 (10%).[56-58]
  • Men with breast cancer (any age): 1 in 20 (5%).[59]
  • Women with ovarian cancer (any age): 1 in 8 to 1 in 10 (10%–15%).[60-62]

Among AJ individuals, the likelihood of having any BRCA pathogenic variant is as follows:

  • General AJ population: 1 in 40 (1.1%–2.5%).[63-65]
  • Women with breast cancer (any age): 1 in 10 (10%).[66]
  • Women with breast cancer (younger than 40 y): 1 in 3 (30%–35%).[66-68]
  • Men with breast cancer (any age): 1 in 5 (19%).[69]
  • Women with ovarian cancer or primary peritoneal cancer (all ages): 1 in 3 (36%–41%).[70-72]

Two large U.S. population-based studies of breast cancer patients younger than 65 years examined the prevalence of BRCA1 [57,73] and BRCA2 [57] pathogenic variants in various ethnic groups. The prevalence of BRCA1 pathogenic variants in breast cancer patients by ethnic group was 3.5% in Hispanic individuals, 1.3% to 1.4% in African American individuals, 0.5% in Asian American individuals, 2.2% to 2.9% in non-AJ White individuals, and 8.3% to 10.2% in AJ individuals.[57,73] The prevalence of BRCA2 pathogenic variants by ethnic group was 2.6% in African American individuals and 2.1% in White individuals.[57]

A study of Hispanic patients with a personal or family history of breast cancer and/or ovarian cancer, who were enrolled through multiple clinics in the southwestern United States, examined the prevalence of BRCA1 and BRCA2 pathogenic variants. BRCA pathogenic variants were identified in 189 of 746 patients (25%) (124 BRCA1, 65 BRCA2);[74] 21 of the 189 (11%) BRCA pathogenic variants identified were large rearrangements, of which 13 (62%) were the BRCA1 exon 9–12 deletion. An unselected cohort of 810 women of Mexican ancestry with breast cancer were tested; 4.3% had a BRCA pathogenic variant. Eight of the 35 pathogenic variants identified also were the BRCA1 exon 9–12 deletion.[75] In another population-based cohort of 492 Hispanic women with breast cancer, the BRCA1 exon 9–12 deletion was found in three patients, suggesting that this variant may be a Mexican founder pathogenic variant and may represent 10% to 12% of all BRCA1 pathogenic variants in similar clinic- and population-based cohorts in the United States. Within the clinic-based cohort, there were nine recurrent pathogenic variants, which accounted for 53% of all variants observed in this cohort, suggesting the existence of additional founder pathogenic variants in this population.

A retrospective review of 29 AJ patients with primary fallopian tube tumors identified germline BRCA pathogenic variants in 17% of patients.[72] Another study of 108 women with fallopian tube cancer identified pathogenic variants in 55.6% of the Jewish women and 26.4% of non-Jewish women (30.6% overall).[76] Estimates of the frequency of fallopian tube cancer in carriers of BRCA pathogenic variants are limited by the lack of precision in the assignment of site of origin for high-grade, metastatic, serous carcinomas at initial presentation.[6,72,76,77]

Population screening

Population screening has identified carriers in a number of AJ populations who would not have met criteria for family-based testing.[64,78-80] This could potentially expand the number of individuals who could benefit from preventive strategies. A study has suggested that population screening (compared with personal/family history–based testing) for AJ founder variants is cost-effective on the basis of data from the United States and the United Kingdom.[81] The authors used a decision-analytic model that estimated lifetime costs and the effects of genetic testing to assess cost-effectiveness; the model included costs of pretest genetic counseling and genetic testing and the anticipated risk of cardiovascular outcomes. Additional analyses conducted by the same group also suggested cost-effectiveness when testing was expanded to include all pathogenic variants in BRCA1, BRCA2, RAD51C, RAD51D, and PALB2.[82] These studies are based on various assumptions, some of which are imprecise (e.g., population prevalence estimates for some genes). Furthermore, as acknowledged by the authors, these types of efforts would require implementation of clinical support across the care continuum, in order for patients identified with pathogenic variants to benefit from this information. Consequently, there remain significant resource implications as population screening efforts are considered, which are the focus of ongoing research efforts. Because the detection rate is highly dependent on the prevalence of pathogenic variants in a population, it is not clear how applicable this approach would be for other populations, including other founder pathogenic variant populations. Another unanswered question is whether adequate genetic counseling can be provided for whole populations.

Clinical criteria and models for prediction of the likelihood of a BRCA1 or BRCA2 pathogenic variant

Several studies have assessed the frequency of BRCA1 or BRCA2 pathogenic variants in women with breast or ovarian cancer.[57,58,73,83-91] Personal characteristics associated with an increased likelihood of a BRCA1 and/or BRCA2 pathogenic variant include the following:

  • Breast cancer diagnosed at an early age. (Some studies use age 40 y as a cutoff, while others use age 50 y.)
  • Ovarian cancer.
  • Bilateral breast cancer.
  • A history of both breast and ovarian cancer.
  • Breast cancer diagnosed in a male at any age.[83-86,89]
  • Triple-negative breast cancer (TNBC) diagnosed in women younger than 60 years.[92-95]
  • AJ background.[83,84,86]

Studies have shown that women with metastatic breast cancer have a higher frequency of inherited pathogenic or likely pathogenic variants in breast cancer risk genes than women with nonmetastatic breast cancer. Mutations were seen in BRCA1, BRCA2, and other breast cancer risk genes. Hence, providers may want to consider genetic testing in women with metastatic breast cancer.[96,97]

Family history characteristics associated with an increased likelihood of carrying a BRCA1 and/or BRCA2 pathogenic variant include the following:

  • Multiple cases of breast cancer.
  • Both breast and ovarian cancer.
  • One or more breast cancers in male family members.
  • AJ background.[83-86]
Clinical criteria and practice guidelines for identifying individuals who may have a BRCA1 or BRCA2 pathogenic variant

Several professional organizations and expert panels, including the American Society of Clinical Oncology,[98] the National Comprehensive Cancer Network (NCCN),[99] the American Society of Human Genetics,[100] the American College of Medical Genetics and Genomics,[101] the National Society of Genetic Counselors,[101] the U.S. Preventive Services Task Force,[102] and the Society of Gynecologic Oncologists,[103] have developed clinical criteria and practice guidelines that can be helpful to health care providers in identifying individuals who may have a BRCA1 or BRCA2 pathogenic variant.

Models for prediction of the likelihood of a BRCA1 or BRCA2 pathogenic variant

Many models have been developed to predict the probability of identifying germline BRCA1/BRCA2 pathogenic variants in individuals or families. These models include those using logistic regression,[32,83,84,86,89,104,105] genetic models using Bayesian analysis (BRCAPRO and Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm [BOADICEA]),[89,106] and empiric observations.[54,57,60,107-109]

In addition to BOADICEA, BRCAPRO is commonly used for genetic counseling in the clinical setting. BRCAPRO and BOADICEA predict the probability of being a carrier and produce estimates of breast cancer risk (refer to Table 4). The discrimination and accuracy (factors used to evaluate the performance of prediction models) of these models are much higher for their ability to report on carrier status than for their ability to predict fixed or remaining lifetime risk.

BOADICEA is a polygenetic model that uses complex segregation analysis to examine both breast cancer risk and the probability of having a BRCA1 or BRCA2 pathogenic variant.[106] Even among experienced providers, the use of prediction models has been shown to increase the power to discriminate which patients are most likely to be carriers of BRCA1/BRCA2 pathogenic variants.[110,111] Most models do not include other cancers seen in the BRCA1 and BRCA2 spectrum, such as pancreatic cancer and prostate cancer. Interventions that decrease the likelihood that an individual will develop cancer (such as oophorectomy and mastectomy) may influence the ability to predict BRCA1 and BRCA2 pathogenic variant status.[112] One study has shown that the prediction models for genetic risk are sensitive to the amount of family history data available and do not perform as well with limited family information.[113] BOADICEA is being expanded to incorporate additional risk variants (genome-wide association study [GWAS] single nucleotide variants [SNVs]) to better predict pathogenic variant status and to improve the accuracy of breast cancer and ovarian cancer risk estimates.[114]

The performance of the models can vary in specific ethnic groups. The BRCAPRO model appeared to best fit a series of French Canadian families.[115] There have been variable results in the performance of the BRCAPRO model among Hispanic individuals,[116,117] and both the BRCAPRO model and Myriad tables underestimated the proportion of carriers of pathogenic variants in an Asian American population.[118] BOADICEA was developed and validated in British women. Thus, the major models used for both overall risk (Table 1) and genetic risk (Table 4) have not been developed or validated in large populations of racially and ethnically diverse women. Of the commonly used clinical models for assessing genetic risk, only the Tyrer-Cuzick model contains nongenetic risk factors.

The power of several of the models has been compared in different studies.[119-122] Four breast cancer genetic-risk models, BOADICEA, BRCAPRO, IBIS, and eCLAUS, were evaluated for their diagnostic accuracy in predicting BRCA1/BRCA2 pathogenic variants in a cohort of 7,352 German families.[123] The family member with the highest likelihood of carrying a pathogenic variant from each family was screened for BRCA1/BRCA2 pathogenic variants. Carrier probabilities from each model were calculated and compared with the actual variants detected. BRCAPRO and BOADICEA had significantly higher diagnostic accuracy than IBIS or eCLAUS. Accuracy for the BOADICEA model was further improved when statuses of the tumor markers ER, progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/neu) were included in the model. The inclusion of these biomarkers has been shown to improve the performance of BRCAPRO.[124,125]

Table 4. Characteristics of Common Models for Estimating the Likelihood of a BRCA1/BRCA2 Pathogenic Variant
  Myriad Prevalence Tables [86] BRCAPRO [89,112] BOADICEA [89,106] Tyrer-Cuzick [126]
AJ = Ashkenazi Jewish; BOADICEA = Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm; FDR = first-degree relatives; SDR = second-degree relatives.
Method Empiric data from Myriad Genetics based on personal and family history reported on requisition forms Statistical model, assumes autosomal dominant inheritance Statistical model, assumes polygenic risk Statistical model, assumes autosomal dominant inheritance
Features of the model Proband may or may not have breast or ovarian cancer Proband may or may not have breast or ovarian cancer Proband may or may not have breast or ovarian cancer Proband must be unaffected
Considers age of breast cancer diagnosis as <50 y, >50 y Considers exact age at breast and ovarian cancer diagnosis Considers exact age at breast and ovarian cancer diagnosis Also includes reproductive factors and body mass index to estimate breast cancer risk
Considers breast cancer in ≥1 affected relative only if diagnosed <50 y Considers prior genetic testing in family (i.e., BRCA1/BRCA2 pathogenic variant–negative relatives) Includes all FDR and SDR with and without cancer  
Considers ovarian cancer in ≥1 relative at any age Considers oophorectomy status Includes AJ ancestry  
Includes AJ ancestry Includes all FDR and SDR with and without cancer    
Very easy to use Includes AJ ancestry    
Limitations Simplified/limited consideration of family structure Requires computer software and time-consuming data entry Requires computer software and time-consuming data entry Designed for individuals unaffected with breast cancer
Incorporates only FDR and SDR; may need to change proband to best capture risk and to account for disease in the paternal lineage
May overestimate risk in bilateral breast cancer [127]
Early age of breast cancer onset May perform better in White populations than in racial and ethnic minority populations [117,128] Incorporates only FDR and SDR; may need to change proband to best capture risk
May underestimate risk of BRCA pathogenic variant in high-grade serous ovarian cancers but overestimate the risk for other histologies [129]

Genetic testing for BRCA1 and BRCA2 pathogenic variants has been available to the public since 1996. As more individuals have undergone testing, risk assessment models have improved. This, in turn, gives providers better data to estimate an individual patient’s risk of carrying a pathogenic variant, but risk assessment continues to be an art. There are factors that might limit the ability to provide an accurate risk assessment (i.e., small family size, paucity of women, or ethnicity) including the specific circumstances of the individual patient (such as history of disease or risk-reducing surgeries).

Penetrance of BRCA pathogenic variants

The proportion of individuals carrying a pathogenic variant who will manifest the disease is referred to as penetrance. (Refer to the Penetrance of Inherited Susceptibility to Hereditary Breast and/or Gynecologic Cancers section of this summary for more information.)

Numerous studies have estimated breast and ovarian cancer penetrance in carriers of BRCA1 and BRCA2 pathogenic variants. Risk of both breast and ovarian cancer is consistently estimated to be higher in carriers of BRCA1 pathogenic variants than in carriers of BRCA2 pathogenic variants. Results from two large meta-analyses are shown in Table 5.[130,131] One study [130] analyzed pooled pedigree data from 22 studies involving 289 BRCA1 and 221 BRCA2 pathogenic variant–positive individuals. Index cases from these studies had female breast cancer, male breast cancer, or ovarian cancer but were unselected for family history. A subsequent study [131] combined penetrance estimates from the previous study and nine others that included an additional 734 BRCA1 and 400 BRCA2 pathogenic variant–positive families. The estimated cumulative risks of breast cancer by age 70 years in these two meta-analyses were 55% to 65% for carriers of BRCA1 pathogenic variants and 45% to 47% for carriers of BRCA2 pathogenic variants. Ovarian cancer risks were 39% for carriers of BRCA1 pathogenic variants and 11% to 17% for carriers of BRCA2 pathogenic variants.

Table 5. Estimated Cumulative Breast and Ovarian Cancer Risks in Carriers of BRCA1 and BRCA2 Pathogenic Variants
Study Breast Cancer Risk (%) (95% CI) Ovarian Cancer Risk (%) (95% CI)
CI = confidence interval.
aRisk estimate calculated up to age 70 years.
bRisk estimate calculated up to age 80 years.
  BRCA1 BRCA2 BRCA1 BRCA2
Antoniou et al. (2003) [130] 65 (44–78)a 45 (31–56)a 39 (18–54)a 11 (2.4–19)a
Chen et al. (2007) [131] 55 (50–59)a 47 (42–51)a 39 (34–45)a 17 (13–21)a
Kuchenbaecker et al. (2017) [132] 72 (65–79)b 69 (61–77)b 44 (36–53)b 17 (11–25)b

While the cumulative risks of developing cancer by age 70 years are higher for carriers of BRCA1 pathogenic variants than for BRCA2 pathogenic variants, the relative risks (RRs) of breast cancer decline more with age in carriers of BRCA1 pathogenic variants.[130] Studies of penetrance for carriers of specific individual variants are not usually large enough to provide stable estimates, but numerous studies of the Ashkenazi founder pathogenic variants have been conducted. One group of researchers analyzed the subset of families with one of the Ashkenazi founder pathogenic variants from their larger meta-analyses and found that the estimated penetrance for the individual pathogenic variants was very similar to the corresponding estimates among all carriers.[133] A later study of 4,649 women with BRCA pathogenic variants reported significantly lower RRs of breast cancer in those with the BRCA2 6174delT variant than in those with other BRCA2 variants (hazard ratio [HR], 0.35; confidence interval [CI], 0.18–0.69).[134]

One study provided prospective 10-year risks of developing cancer among asymptomatic carriers at various ages.[131] Nonetheless, making precise penetrance estimates in an individual carrier is difficult. The lifetime risks of ovarian cancer are 5.2% in carriers of RAD51C pathogenic variants, 5.8% in carriers of BRIP1 pathogenic variants, and 12% in carriers of RAD51D pathogenic variants. Risk-reducing salpingo-oophorectomy (RRSO) may be considered for these patients upon completion of childbearing.[135,136]

Data from the Consortium of Investigators of Modifiers of BRCA1/BRCA2 (CIMBA), comprising 19,581 carriers of BRCA1 pathogenic variants and 11,900 carriers of BRCA2 pathogenic variants, were analyzed to estimate HRs for breast cancer and ovarian cancer by pathogenic variant type, function, and nucleotide position.[137] Breast cancer cluster regions and ovarian cancer cluster regions were found in both genes. Risks for incidence of breast cancer and ovarian cancer and age at diagnosis differed by variant class. Further evaluation of these findings is needed before they can be translated into clinical practice.

Another study from the CIMBA group looked at the phenotype of women with breast cancer who had inherited pathogenic variants in both BRCA1 and BRCA2.[138] The majority of women carried the common Jewish pathogenic variants. Compared with women who were heterozygous for the same pathogenic variant (heterozygote controls), women who were heterozygous for both BRCA1 and BRCA2 were more likely to be diagnosed with breast cancer than women who were heterozygote controls, and more likely to be diagnosed with ovarian cancer than women who were heterozygote controls with BRCA2, but not those with BRCA1 pathogenic variants. Similarly, age at onset of breast cancer was younger in carriers of both variants compared with women who were heterozygote controls with BRCA2, but not compared with those with BRCA1 pathogenic variants. The percentage of women with both variants and estrogen receptor–positive and progesterone receptor–positive breast cancer was intermediate between the heterozygote controls with BRCA1 pathogenic variants and those with BRCA2 pathogenic variants. The authors concluded that women who inherit pathogenic variants in both BRCA1 and BRCA2 may be managed similarly to carriers of only a BRCA1 variant.

Several studies have suggested that BRCA pathogenic variants may be associated with genetic anticipation. One study evaluated 176 families with BRCA1 or BRCA2 pathogenic variants and at least two consecutive generations of the same cancer. The probands’ generations were diagnosed with breast cancer an estimated 6.8 years earlier than the parents’ generations and 9.8 years earlier than the grandparents' generations.[139] Similarly, another study showed a difference in age at breast cancer diagnosis between 80 mother-and-daughter paired pathogenic variant carriers but only if the mother was diagnosed with breast cancer after age 50 years.[140] Another cohort study of 106 paired women from two consecutive generations with a known BRCA pathogenic variant in the family estimated a 6- to 8-year earlier age at onset in subsequent generations.[141]

RRSO and/or use of oral contraceptives have been associated with the risk of breast cancer.[66,130,142-147] (Refer to the RRSO section and the Oral contraceptives section of this summary for more information.) Other potentially modifiable reproductive and hormonal factors can also affect risk.[148-152] Genetic modifiers of penetrance of breast cancer and ovarian cancer are increasingly under study but are not clinically useful at this time.[153-155] (Refer to the Modifiers of risk in carriers of BRCA1 and BRCA2 pathogenic variants section for more information.) While the average breast cancer and ovarian cancer penetrances may not be as high as initially estimated, they are substantial, both in relative and absolute terms, particularly in women born after 1940. A higher risk before age 50 years has been consistently seen in more recent birth cohorts,[64,66,141] and additional studies will be required to further characterize potential modifying factors to arrive at more precise individual risk projections. Precise penetrance estimates for less common cancers, such as pancreatic cancer, are lacking.

Contralateral breast cancer (CBC) in carriers of BRCA pathogenic variants

The increased risk of CBC among carriers of BRCA1 and BRCA2 pathogenic variants has been confirmed in several large studies, with fairly consistent results, as summarized in Table 6.

Table 6. Contralateral Breast Cancer 10-Year Cumulative Risk Estimates for Carriers of BRCA1/BRCA2 Pathogenic Variants
Study BRCA1 Carriers (%) BRCA2 Carriers (%)
Graeser et al. (2009) [156] 18.5 13.2
Malone et al. (2010) [157] 20.5 15.9
van der Kolk et al. (2010) [158] 34.2 29.2
Metcalfe et al. (2011) [159] 23.8 18.7
Molina-Montes et al. (2014) [160] 27 19
Basu et al. (2015) [161] 25.7 19.5
van den Broek et al. (2016) [162] 21.1 10.8

Published results include a large study by the German Consortium for Hereditary Breast and Ovarian Cancer, which estimated the risk of CBC in members of families with known BRCA1 and BRCA2 pathogenic variants. At 25 years after the first breast cancer, the risk of CBC was close to 50% in both BRCA1 and BRCA2 families. The risk was also inversely correlated with age in this study, with the highest risks seen in women whose first breast cancer was before age 40 years.[156]

Subsequently, results from the Women's Environmental Cancer and Radiation Epidemiology (WECARE) study, a large, population-based, nested case-control study of CBC, reported a 10-year risk of CBC of 15.9% among carriers of BRCA1/BRCA2 pathogenic variants and a risk of 4.9% among noncarriers. Risks were also inversely related to age at first diagnosis in this study and were 1.8-fold higher in those with a first-degree relative (FDR) with breast cancer.[157]

A larger study of members of BRCA1/BRCA2 families in the Netherlands reported similar 10-year risks of CBC for women from BRCA1 and BRCA2 families (34.2% and 29.2%, respectively).[158]

A comparison of 655 women with BRCA1/BRCA2 pathogenic variants undergoing either breast-conserving therapy or mastectomy noted that both treatment groups experienced high rates of CBC, exceeding 50% by 20 years of follow-up. Rates were significantly higher among women with BRCA1 pathogenic variants than in women with BRCA2 pathogenic variants, and among women whose first breast cancer occurred at or before age 35 years.[163]

In a study of 810 women with stage I or stage II breast cancer who had a BRCA1 or BRCA2 pathogenic variant identified in the family, 149 (18.4%) developed CBC; the 15-year actuarial risk was 36.1% among carriers of BRCA1 pathogenic variants and 28.5% among carriers of BRCA2 pathogenic variants.[159] Risks were higher among women diagnosed before age 50 years than among women diagnosed at age 50 years or older (37.6% vs. 16.8%; P = .003). Furthermore, the risk of CBC varied by family history among women whose initial breast cancer was diagnosed before age 50 years. For these women, the CBC risk among those with 0, 1, or 2 or more FDRs with breast cancer diagnosed before age 50 years was 33.4%, 39.1%, and 49.7%, respectively.

The risk of CBC after a first breast cancer in BRCA1 and BRCA2 carriers has been examined in both retrospective and prospective observational epidemiological studies. A systematic review and quantitative meta-analysis of these epidemiologic studies (18 retrospective and 2 prospective cohort studies) reported 5-year cumulative risks of CBC of 15% (95% CI, 9.50%–20%) in BRCA1 carriers and 9% (95% CI, 5%–14%) in BRCA2 carriers.[160] When the prospective studies were analyzed separately, the 5-year cumulative risk increased to 23.4% (95% CI, 9.1%–39.5%) in BRCA1 carriers and to 17.5% (95% CI, 9.1%–39.5%) in BRCA2 carriers. The discrepancies in the reported frequencies may be inherent due to the potential for biases introduced in retrospective series.

Similarly, in a Dutch cohort of 6,294 patients (including 200 BRCA1 carriers and 71 BRCA2 carriers) with invasive breast cancer diagnosed before age 50 years, and a median follow-up of 12.5 years, the 10-year risks of CBC were 21.1% (95% CI, 15.4%–27.4%) for BRCA1 carriers, 10.8% (95% CI, 4.7%–19.6%) for BRCA2 carriers, and 5.1% (95% CI, 4.5%–5.7%) for noncarriers.[162] Age at first breast cancer diagnosis was predictive of the 10-year cumulative risk of CBC among BRCA1/BRCA2 carriers only. Specifically, the CBC risk among BRCA1/BRCA2 carriers diagnosed before age 41 years was 23.9% (BRCA1, 25.5%; BRCA2, 17.2%); in contrast, CBC among those diagnosed between ages 41 and 49 years was 12.6% (BRCA1, 15.6%; BRCA2, 7.2%).

In an English study of 506 BRCA1 carriers and 505 BRCA2 carriers with a diagnosis of breast cancer at any age and median follow-up of 7.8 years, the 10-year risks for CBC were 25.7% for BRCA1 carriers and 19.5% for BRCA2 carriers.[161] Earlier age at first breast cancer diagnosis for BRCA1 and BRCA2 carriers combined was significantly associated with a higher CBC risk, with a 20-year rate of 55.4% among those younger than 40 years, compared with 36.4% among those older than 50 years. Additionally, differences were more pronounced among BRCA1 carriers when compared with BRCA2 carriers.

An international, multicenter, prospective cohort study followed 1,305 BRCA1 and 908 BRCA2 female carriers with a diagnosis of breast cancer (without any other cancers) for a median follow-up time of 4 years (range, 2–7 y).[132] Participants had a median age of 47 years (range, 40–55 y) at the start of follow-up. The authors reported a cumulative risk of CBC 20 years after the initial breast cancer diagnosis of 40% (95% CI, 35%–45%) for BRCA1 carriers and 26% (95% CI, 20%–33%) for BRCA2 carriers. These 20-year estimates are comparable to 10-year cumulative risk estimates reported in Table 6.

The effect of radiation therapy on CBC risk has also been investigated. A population-based case-control study of 708 women with CBC and 1,399 controls with unilateral breast cancer showed no clear association between the use of radiation therapy and an increased risk of CBC in patients with BRCA1/BRCA2 pathogenic variants.[164] There was no evidence of increased CBC risk following radiation use in carriers of pathogenic/likely pathogenic ATM variants or in carriers of the pathogenic 1100delC CHEK2 variant.

Thus, in summary, despite differences in study design, study sites, and sample sizes, the data on CBC among women with BRCA1/BRCA2 pathogenic variants show several consistent findings:

  • The risk at all time points studied is significantly higher than that among sporadic controls.
  • The risk continues to rise with time since an individual's first breast cancer. Risk reaches 20% to 30% at 10 years of follow-up, and 40% to 50% at 20 years of follow-up in most studies.
  • Some, but not all, studies show an excess of CBC among BRCA1 carriers when compared with BRCA2 carriers.
  • The risk of CBC is greatest among women whose first breast cancer occurs at a young age, and thus is inversely associated with age.
Risk-reducing strategies

Refer to the Risk-reducing mastectomy section of this summary for information about the use of risk-reducing surgery in carriers of BRCA pathogenic variants. Refer to the Chemoprevention section of this summary for information about the use of tamoxifen as a risk-reduction strategy for CBC in carriers of BRCA pathogenic variants.

Breast cancer as a second malignancy in carriers of BRCA pathogenic variants

Two genetic registry–based studies have recently explored the risk of primary breast cancer after BRCA-related ovarian cancer. In one study, 164 BRCA1/BRCA2 carriers with primary epithelial ovarian, fallopian tube or primary peritoneal cancer were followed for subsequent events.[165] The risk of metachronous breast cancer at 5 years after a diagnosis of ovarian cancer was lower than previously reported for unaffected BRCA1/BRCA2 carriers. In this series, overall survival was dominated by ovarian cancer-related deaths. A similar study compared the risk of primary breast cancer in BRCA-related ovarian cancer patients and unaffected carriers.[166] The 2-year, 5-year, and 10-year risks of primary breast cancer were all statistically significantly lower in patients with ovarian cancer. The risk of CBC among women with a unilateral breast cancer before their ovarian cancer diagnosis was also lower than in women without ovarian cancer, although the difference did not reach statistical significance. These studies suggest that treatment for ovarian cancer, namely oophorectomy and platinum-based chemotherapy, may confer protection against subsequent breast cancer. In a single-institution cohort study of 364 patients with epithelial ovarian cancer who underwent BRCA pathogenic variant testing, 135 (37.1%) were found to carry a germline BRCA1 or BRCA2 pathogenic variant. Of the 135 BRCA1/BRCA2 carriers, 12 (8.9%) developed breast cancer. All breast cancers were stage 0 to stage II and diagnosed as follows: mammogram (7), palpable mass (3), and incidental finding during risk-reducing mastectomy (2). At median follow-up of 6.3 years, of the 12 patients with breast cancer after ovarian cancer, three died of recurrent ovarian cancer and one died of metastatic breast cancer.[167] The majority of these cancers were detected with mammogram or clinical exam, suggesting additional breast surveillance with other modalities or risk-reducing surgery may be of questionable value. Mathematical modeling suggests that for women with BRCA-associated ovarian cancer, breast cancer screening should consist of mammography and clinical breast exam. The consideration of breast magnetic resonance imaging (MRI) and/or risk-reducing mastectomies may be beneficial for women with early-stage ovarian cancer or for long-term ovarian cancer survivors.[168]

Cancers other than female breast/ovarian

Female breast and ovarian cancers are clearly the dominant cancers associated with BRCA1 and BRCA2. BRCA pathogenic variants also confer an increased risk of fallopian tube and primary peritoneal carcinomas. One large study from a familial registry of carriers of BRCA1 pathogenic variants has found a 120-fold RR of fallopian tube cancer among carriers of BRCA1 pathogenic variants compared with the general population.[6] The risk of primary peritoneal cancer among carriers of BRCA pathogenic variants with intact ovaries is increased but remains poorly quantified, despite a residual risk of 3% to 4% in the 20 years after RRSO.[169,170] (Refer to the RRSO section in the Ovarian cancer section of this summary for more information.)

Pancreatic, male breast, and prostate cancers have also been consistently associated with BRCA pathogenic variants, particularly with BRCA2. Other cancers have been associated in some studies. The strength of the association of these cancers with BRCA pathogenic variants has been more difficult to estimate because of the lower numbers of these cancers observed in carriers of pathogenic variants.

Men with BRCA2 pathogenic variants, and to a lesser extent BRCA1 pathogenic variants, are at increased risk of breast cancer with lifetime risks estimated at 5% to 10% and 1% to 2%, respectively.[6,8,9,171] Men carrying BRCA2 pathogenic variants, and to a lesser extent BRCA1 pathogenic variants, have an approximately threefold to sevenfold increased risk of prostate cancer.[7,8,12,109,172-175] BRCA2-associated prostate cancer also appears to be more aggressive.[176-181] (Refer to the BRCA1 and BRCA2 section in the PDQ summary on Genetics of Prostate Cancer for more information.)

Studies of familial pancreatic cancer (FPC) [182-186] and unselected series of pancreatic cancer [187-189] have also supported an association with BRCA2, and to a lesser extent, BRCA1.[7] Overall, it appears that between 3% to 15% of families with FPC may have germline BRCA2 pathogenic variants, with risks increasing with more affected relatives.[182-184] Similarly, studies of unselected pancreatic cancers have reported BRCA2 pathogenic variant frequencies between 3% to 7%, with these numbers approaching 10% in those of AJ descent.[187,188,190] The lifetime risk of pancreatic cancer in BRCA2 carriers is estimated to be 3% to 5%,[8,12] compared with an estimated lifetime risk of 0.5% by age 70 years in the general population.[191] A large, single-institution study of more than 1,000 carriers of pathogenic variants found a 21-fold increased risk of pancreatic cancer among BRCA2 carriers and a 4.7-fold increased risk among carriers of BRCA1 pathogenic variants, compared with incidence in the general population.[175] Other cancers associated with BRCA2 pathogenic variants in some, but not all, studies include melanoma, biliary cancers, and head and neck cancers, but these risks appear modest (<5% lifetime risk) and are less well studied.[12]

Table 7. Spectrum of Cancers in Carriers of BRCA1 and BRCA2 Pathogenic Variants
Cancer Sites [6-8,12,63,174] BRCA1 BRCA2
aRefer to the PDQ summary on Genetics of Prostate Cancer for more information about the association of BRCA1 and BRCA2 with prostate cancer.
+++ Multiple studies demonstrated association and are relatively consistent.
++ Multiple studies and the predominance of the evidence are positive.
+ May be an association, predominantly single studies; smaller limited studies and/or inconsistent but weighted toward positive.
  Strength of Evidence Magnitude of Absolute Risk Strength of Evidence Magnitude of Absolute Risk
Breast (female) +++ High +++ High
Ovary, fallopian tube, peritoneum +++ High +++ Moderate
Breast (male) + Undefined +++ Low
Pancreas ++ Very Low +++ Low
Prostatea + Undefined +++ High

The first Breast Cancer Linkage Consortium study investigating cancer risks reported an excess of colorectal cancer in BRCA1 carriers (RR, 4.1; 95% CI, 2.4–7.2).[192] This finding was supported by some,[6,7,193] but not all,[8,63,71,109,194-196] family-based studies. However, unselected series of colorectal cancer that have been exclusively performed in the AJ population have not shown elevated rates of BRCA1 or BRCA2 pathogenic variants.[197-199] Taken together, the data suggest little, if any, increased risk of colorectal cancer, and possibly only in specific population groups. Therefore, at this time, carriers of BRCA1 pathogenic variants should adhere to population-screening recommendations for colorectal cancer.

No increased prevalence of hereditary BRCA pathogenic variants was found among 200 Jewish women with endometrial carcinoma or 56 unselected women with uterine papillary serous carcinoma.[200,201] (Refer to the Risk-reducing salpingo-oophorectomy section in the Ovarian cancer section of this summary for more information.)

Cancer risk in individuals who test negative for a known familial BRCA1/BRCA2 pathogenic variant ("true negative")

There is conflicting evidence as to the residual familial risk among women who test negative for the BRCA1/BRCA2 pathogenic variant segregating in the family. An initial study based on prospective evaluation of 353 women who tested negative for the BRCA1 pathogenic variant segregating in the family found that five incident breast cancers occurred during more than 6,000 person-years of observation, for a lifetime risk of 6.8%, a rate similar to the general population.[145] A report that the risk may be as high as fivefold in women who tested negative for the BRCA1 or BRCA2 pathogenic variant in the family [202] was followed by numerous letters to the editor suggesting that ascertainment biases account for much of this observed excess risk.[203-208] Four additional analyses have suggested an approximate 1.5-fold to 2-fold excess risk.[207,209-211] In one study, two cases of ovarian cancer were reported.[211] Several studies have involved retrospective analyses; all studies have been based on small observed numbers of cases and have been of uncertain statistical and clinical significance.

Results from numerous other prospective studies have found no increased risk. A study of 375 women who tested negative for a known familial pathogenic variant in BRCA1 or BRCA2 reported two invasive breast cancers, two in situ breast cancers, and no ovarian cancers diagnosed, with a mean follow-up of 4.9 years. Four invasive breast cancers were expected, whereas two were observed.[212] Another study of similar size but longer follow-up (395 women and 7,008 person-years of follow-up) also found no statistically significant overall increase in breast cancer risk among variant-negative women (observed/expected [O/E], 0.82; 95% CI, 0.39–1.51), although women who had at least one FDR with breast cancer had a nonsignificant increased risk (O/E, 1.33; 95% CI, 0.41–2.91).[213] A study of 160 BRCA1 and 132 BRCA2 pathogenic variant–positive families from the Breast Cancer Family Registry found no evidence for increased risk among noncarriers in these families.[214] In a large study of 722 variant-negative women from Australia in whom six invasive breast cancers were observed after a median follow-up of 6.3 years, the standardized incidence ratio (SIR) was not significantly elevated (SIR, 1.14; 95% CI, 0.51–2.53).[215] Based on available data, it appears that women testing negative for known familial BRCA1/BRCA2 pathogenic variants can adhere to general population screening guidelines unless they have sufficient additional risk factors, such as a personal history of atypical hyperplasia of the breast or family history of breast cancer in relatives who do not carry the familial pathogenic variant.

Breast and ovarian cancer risk in breast cancer families without detectable BRCA1/BRCA2 pathogenic variants ("indeterminate")

The majority of families with site-specific breast cancer test negative for BRCA1/BRCA2 and have no features consistent with Cowden syndrome or Li-Fraumeni syndrome.[32] Five studies using population-based and clinic-based approaches have demonstrated no increased risk of ovarian cancer in such families. Although ovarian cancer risk was not increased, breast cancer risk remained elevated.[214,216,216,217,217,218,218-220]

Modifiers of risk in carriers of BRCA1 and BRCA2 pathogenic variants

Pathogenic variants in BRCA1 and BRCA2 confer high risks of breast and ovarian cancers. The risks, however, are not equal in all pathogenic variant carriers and have been found to vary by several factors, including type of cancer, age at onset, and variant position.[221] This observed variation in penetrance has led to the hypothesis that other genetic and/or environmental factors modify cancer risk in carriers of pathogenic variants. There is a growing body of literature identifying genetic and nongenetic factors that contribute to the observed variation in rates of cancers seen in families with BRCA1/BRCA2 pathogenic variants.

Genetic modifiers of breast and ovarian cancer risk

The largest studies investigating genetic modifiers of breast and ovarian cancer risk to date have come from CIMBA, a large international effort with genotypic and phenotypic data on more than 15,000 BRCA1 and 10,000 BRCA2 carriers.[222] Using candidate gene analysis and GWAS, CIMBA has identified several loci associated both with increased and decreased risk of breast cancer and ovarian cancer. Some of the SNVs are related to subtypes of breast cancer, such as hormone-receptor and HER2/neu status. The risks conferred are all modest but if operating in a multiplicative fashion could significantly impact risk of cancer in carriers of BRCA1/BRCA2 pathogenic variants. Currently, these SNVs are not being tested for or used in clinical decision making.

Genotype-phenotype correlations

Some genotype-phenotype correlations have been identified in both BRCA1 and BRCA2 pathogenic variant families. None of the studies have had sufficient numbers of pathogenic variant–positive individuals to make definitive conclusions, and the findings are probably not sufficiently established to use in individual risk assessment and management. In 25 families with BRCA2 pathogenic variants, an ovarian cancer cluster region was identified in exon 11 bordered by nucleotides 3,035 and 6,629.[11,223] A study of 164 families with BRCA2 pathogenic variants collected by the Breast Cancer Linkage Consortium confirmed the initial finding. Pathogenic variants within the ovarian cancer cluster region were associated with an increased risk of ovarian cancer and a decreased risk of breast cancer in comparison with families with variants on either side of this region.[224] In addition, a study of 356 families with protein-truncating BRCA1 pathogenic variants collected by the Breast Cancer Linkage Consortium reported breast cancer risk to be lower with variants in the central region (nucleotides 2,401–4,190) compared with surrounding regions. Ovarian cancer risk was significantly reduced with variants 3’ to nucleotide 4,191.[225] These observations have generally been confirmed in subsequent studies.[130,226,227] Studies in Ashkenazim, in whom substantial numbers of families with the same pathogenic variant can be studied, have also found higher rates of ovarian cancer in carriers of the BRCA1:185delAG variant, in the 5' end of BRCA1, compared with carriers of the BRCA1:5382insC variant in the 3' end of the gene.[228,229] The risk of breast cancer, particularly bilateral breast cancer, and the occurrence of both breast and ovarian cancer in the same individual, however, appear to be higher in carriers of the BRCA1:5382insC pathogenic variant compared with carriers of BRCA1:185delAG and BRCA2:6174delT variants. Ovarian cancer risk is considerably higher in carriers of BRCA1 pathogenic variants, and it is uncommon before age 45 years in carriers of the BRCA2:6174delT pathogenic variant.[228,229]

In an Australian study of 122 families with a pathogenic variant in BRCA1, large genomic rearrangement variants were associated with higher-risk features in breast and ovarian cancers, including younger age at breast cancer diagnosis and higher incidence of bilateral breast cancer.[230]

Pathology of breast cancer

BRCA1 pathology

Several studies evaluating pathologic patterns seen in BRCA1-associated breast cancers have suggested an association with adverse pathologic and biologic features. These findings include higher than expected frequencies of medullary histology, high histologic grade, areas of necrosis, trabecular growth pattern, aneuploidy, high S-phase fraction, high mitotic index, and frequent TP53 variants.[231-238] In a large international series of 3,797 carriers of BRCA1 pathogenic variants, the median age at breast cancer diagnosis was 40 years.[238] Of breast tumors arising in BRCA1 carriers, 78% were ER-negative; 79% were PR-negative; 90% were HER2-negative; and 69% were triple-negative. These findings were consistent with multiple smaller series.[92,234,239-241] In addition, the proportion of ER-negative tumors significantly decreased as the age at breast cancer diagnosis increased.[238]

There is considerable, but not complete, overlap between the triple-negative and basal-like subtype cancers, both of which are common in BRCA1-associated breast cancer,[242,243] particularly in women diagnosed before age 50 years.[92-94] A small proportion of BRCA1-related breast cancers are ER-positive, which are associated with later age of onset.[244,245] These ER-positive cancers have clinical behavior features that are intermediate between ER-negative BRCA1 cancers and ER-positive sporadic breast cancers, raising the possibility that there may be a unique mechanism by which they develop.

The prevalence of germline BRCA1 pathogenic variants in women with TNBC is significant, both in women undergoing clinical genetic testing (and thus selected in large part for family history) and in unselected triple-negative patients, with pathogenic variants reported in 9% to 35%.[94,95,239,246-249] Notably, studies have demonstrated a high rate of BRCA1 pathogenic variants in unselected women with TNBC, particularly in those diagnosed before age 50 years. A large report of 1,824 patients with TNBC unselected for family history, recruited through 12 studies, identified 14.6% with a pathogenic variant in an inherited cancer susceptibility gene.[249] BRCA1 pathogenic variants accounted for the largest proportion (8.5%), followed by BRCA2 (2.7%); PALB2 (1.2%); and BARD1, RAD51D, RAD51C and BRIP1 (0.3%–0.5% for each gene). In this study, those with pathogenic variants in BRCA1/BRCA2 or other inherited cancer genes were diagnosed at an earlier age and had higher grade tumors than those without pathogenic variants. Specifically, among carriers of BRCA1 pathogenic variants, the average age at diagnosis was 44 years, and 94% had high-grade tumors. One study examined 308 individuals with TNBC; BRCA1 pathogenic variants were present in 45. Pathogenic variants were seen both in women unselected for family history (11 of 58; 19%) and in those with family history (26 of 111; 23%).[250] A meta-analysis based on 2,533 patients from 12 studies was conducted to assess the risk of a BRCA1 pathogenic variant in high-risk women with TNBC.[251] Results indicated that the RR of a BRCA1 pathogenic variant among women with versus without TNBC is 5.65 (95% CI, 4.15–7.69), and approximately two in nine women with triple-negative disease harbor a BRCA1 pathogenic variant. Interestingly, a study of 77 unselected patients with TNBC in which 15 (19.5%) had a germline pathogenic variant or somatic BRCA1/BRCA2 mutation demonstrated a lower risk of relapse in those with BRCA1 pathogenic variant–associated TNBC than in those with non-BRCA1-associated TNBC; this study was limited by its size.[247] A second study examining clinical outcomes in BRCA1-associated versus non-BRCA1-associated TNBC showed no difference, although there was a trend toward more brain metastases in those with BRCA1-associated breast cancer. In both of these studies, all but one carrier of BRCA1 pathogenic variants received chemotherapy.[252] In contrast, HER2 positivity and young age alone in the absence of family history or a second primary cancer does not increase the likelihood of a pathogenic variant in BRCA1, BRCA2, or TP53.[253]

It has been hypothesized that many BRCA1 tumors are derived from the basal epithelial layer of cells of the normal mammary gland, which account for 3% to 15% of unselected invasive ductal cancers. If the basal epithelial cells of the breast represent the breast stem cells, the regulatory role suggested for wild-type BRCA1 may partly explain the aggressive phenotype of BRCA1-associated breast cancer when BRCA1 function is damaged.[254] Further studies are needed to fully appreciate the significance of this subtype of breast cancer within the hereditary syndromes.

The most accurate method for identifying basal-like breast cancers is through gene expression studies, which have been used to classify breast cancers into biologically and clinically meaningful groups.[240,255,256] This technology has also been shown to correctly differentiate BRCA1- and BRCA2-associated tumors from sporadic tumors in a high proportion of cases.[257-259] Notably, among a set of breast tumors studied by gene expression array to determine molecular phenotype, all tumors with BRCA1 alterations fell within the basal tumor subtype;[240] however, this technology is not in routine use due to its high cost. Instead, immunohistochemical markers of basal epithelium have been proposed to identify basal-like breast cancers, which are typically negative for ER, PR, and HER2, and stain positive for cytokeratin 5/6, or epidermal growth factor receptor.[260-263] Based on these methods to measure protein expression, a number of studies have shown that the majority of BRCA1-associated breast cancers are positive for basal epithelial markers.[92,234,262]

There is growing evidence that preinvasive lesions are a component of the BRCA phenotype. The Breast Cancer Linkage Consortium initially reported a relative lack of an in situ component in BRCA1-associated breast cancers,[232] also seen in two subsequent studies of BRCA1/BRCA2 carriers.[264,265] However, in a study of 369 ductal carcinoma in situ (DCIS) cases, BRCA1 and BRCA2 pathogenic variants were detected in 0.8% and 2.4%, respectively, which is only slightly lower than previously reported prevalence in studies of invasive breast cancer patients.[266] A retrospective study of breast cancer cases in a high-risk clinic found similar rates of preinvasive lesions, particularly DCIS, among 73 BRCA-associated breast cancers and 146 pathogenic variant–negative cases.[267,268] A study of AJ women, stratified by whether they were referred to a high-risk clinic or were unselected, showed similar prevalence of DCIS and invasive breast cancers in referred patients compared with one-third lower DCIS cases among unselected subjects.[269] Similarly, data about the prevalence of hyperplastic lesions have been inconsistent, with reports of increased [270,271] and decreased prevalence.[265] Similar to invasive breast cancer, DCIS diagnosed at an early age and/or with a family history of breast and/or ovarian cancer is more likely to be associated with a BRCA1/BRCA2 pathogenic variant.[272]

Overall evidence suggests DCIS is part of the BRCA1/BRCA2 spectrum, particularly BRCA2; however, the prevalence of pathogenic variants in DCIS patients, unselected for family history, is less than 5%.[266,269]

BRCA2 pathology

The phenotype for BRCA2-related tumors appears to be more heterogeneous and is less well-characterized than that of BRCA1, although they are generally positive for ER and PR.[232,273,274] A large international series of 2,392 carriers of BRCA2 pathogenic variants found that only 23% of tumors arising in carriers of BRCA2 pathogenic variants were ER-negative; 36% were PR-negative; 87% were HER2-negative; and 16% were triple-negative.[238] A large report of 1,824 patients with TNBC unselected for family history, recruited through 12 studies, identified 2.7% with a BRCA2 pathogenic variant.[249] (Refer to the BRCA1 pathology section of this summary for more information about this study.) A report from Iceland found less tubule formation, more nuclear pleomorphism, and higher mitotic rates in BRCA2-related tumors than in sporadic controls; however, a single BRCA2 founder pathogenic variant (999del5) accounts for nearly all hereditary breast cancer in this population, thus limiting the generalizability of this observation.[275] A large case series from North America and Europe described a greater proportion of BRCA2-associated tumors with continuous pushing margins (a histopathologic description of a pattern of invasion), fewer tubules and lower mitotic counts.[276] Other reports suggest that BRCA2-related tumors include an excess of lobular and tubulolobular histology.[233,273] In summary, histologic characteristics associated with BRCA2 pathogenic variants have been inconsistent.

Role of BRCA1 and BRCA2 in sporadic breast cancer

Given that germline pathogenic variants in BRCA1 or BRCA2 lead to a very high probability of developing breast cancer, it was a natural assumption that these genes would also be involved in the development of the more common nonhereditary forms of the disease. Although somatic mutations in BRCA1 and BRCA2 are not common in sporadic breast cancer tumors,[277-280] there is increasing evidence that hypermethylation of the gene promoter (BRCA1) and loss of heterozygosity (LOH) (BRCA2) are frequent events. In fact, many breast cancers have low levels of the BRCA1 mRNA, which may result from hypermethylation of the gene promoter.[281-283] Approximately 10% to 15% of sporadic breast cancers appear to have BRCA1 promoter hypermethylation, and even more have downregulation of BRCA1 by other mechanisms. Basal-type breast cancers (ER negative, PR negative, HER2 negative, and cytokeratin 5/6 positive) more commonly have BRCA1 dysregulation than other tumor types.[284-286] BRCA1-related tumor characteristics have also been associated with constitutional methylation of the BRCA1 promoter. In a study of 255 breast cancers diagnosed before age 40 years in women without germline BRCA1 pathogenic variants, methylation of BRCA1 in peripheral blood was observed in 31% of women whose tumors had multiple BRCA1-associated pathological characteristics (e.g., high mitotic index and growth pattern including multinucleated cells) compared with less than 4% methylation in controls.[287] (Refer to the BRCA1 pathology section for more information.) Although hypermethylation has not been reported for BRCA2 pathogenic variants, the BRCA2 locus on chromosome 13q is the target of frequent LOH in breast cancer.[288,289] Targeted therapies are being developed for tumors with loss of BRCA1 or BRCA2 protein expression.[290]

Pathology of ovarian cancer

Ovarian cancers in women with BRCA1 and BRCA2 pathogenic variants are more likely to be high-grade serous adenocarcinomas and are less likely to be mucinous or borderline tumors.[291-295] Fallopian tube cancer and peritoneal carcinomas are also part of the BRCA-associated disease spectrum.[72,296]

Histopathologic examinations of fallopian tubes removed from women with a hereditary predisposition to ovarian cancer show dysplastic and hyperplastic lesions that suggest a premalignant phenotype.[297,298] Occult carcinomas have been reported in 2% to 11% of adnexa removed from carriers of BRCA pathogenic variants at the time of risk-reducing surgery.[299-301] Most of these occult lesions are seen in the fallopian tubes, which has led to the hypothesis that many BRCA-associated ovarian cancers may actually have originated in the fallopian tubes. Specifically, the distal segment of the fallopian tubes (containing the fimbriae) has been implicated as a common origin of the high-grade serous cancers seen in BRCA pathogenic variant carriers, based on the close proximity of the fimbriae to the ovarian surface, exposure of the fimbriae to the peritoneal cavity, and the broad surface area in the fimbriae.[302] Because of the multicentric origin of high-grade serous carcinomas from Müllerian-derived tissue, staging of ovarian, tubal, and peritoneal carcinomas is now considered collectively by the International Federation of Gynecology and Obstetrics. The term high-grade serous ovarian carcinoma may be used to represent high-grade pelvic serous carcinoma for consistency in language.[303]

High-grade serous ovarian carcinomas have a higher incidence of somatic TP53 mutations.[291,304] DNA microarray technology suggests distinct molecular pathways of carcinogenesis between BRCA1, BRCA2, and sporadic ovarian cancer.[305] Furthermore, data suggest that BRCA-related ovarian cancers metastasize more frequently to the viscera, while sporadic ovarian cancers remain confined to the peritoneum.[306]

Unlike high-grade serous carcinomas, low-grade serous ovarian cancers are less likely to be part of the BRCA1/BRCA2 spectrum.[307,308]

Role of BRCA1 and BRCA2 in sporadic ovarian cancer

Given that germline variants in BRCA1 or BRCA2 lead to a very high probability of developing ovarian cancer, it was a natural assumption that these genes would also be involved in the development of the more common nonhereditary forms of the disease. Although somatic mutations in BRCA1 and BRCA2 are not common in sporadic ovarian cancer tumors,[277-280] there is increasing evidence that hypermethylation of the gene promoter (BRCA1) and LOH (BRCA2) are frequent events. Loss of BRCA1 or BRCA2 protein expression is more common in ovarian cancer than in breast cancer,[309] and downregulation of BRCA1 is associated with enhanced sensitivity to cisplatin and improved survival in this disease.[310,311] Targeted therapies are being developed for tumors with loss of BRCA1 or BRCA2 protein expression.[290]

Other High-Penetrance Syndromes Associated With Breast and/or Gynecologic Cancers

Lynch syndrome

Lynch syndrome is characterized by autosomal dominant inheritance of susceptibility to predominantly right-sided colon cancer, endometrial cancer, ovarian cancer, and other extracolonic cancers (including cancer of the renal pelvis, ureter, small bowel, and pancreas), multiple primary cancers, and a young age of onset of cancer.[312] The condition is caused by germline variants in the mismatch repair (MMR) genes, which are involved in repair of DNA mismatch variants.[313] The MLH1 and MSH2 genes are the most common susceptibility genes for Lynch syndrome, accounting for 80% to 90% of observed pathogenic variants,[314,315] followed by MSH6 and PMS2.[316-321] (Refer to the Lynch Syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information about this syndrome.)

After colorectal cancer, endometrial cancer is the second hallmark cancer of a family with Lynch syndrome. Even in the original Family G, described by Dr. Aldred Scott Warthin, numerous family members were noted to have extracolonic cancers including endometrial cancer. Although the first version of the Amsterdam criteria did not include endometrial cancer,[322] in 1999, the Amsterdam criteria were revised to include endometrial cancer as extracolonic tumors associated with Lynch syndrome to identify families at risk.[323] In addition, the Bethesda guidelines in 1997 (revised in 2004) did include endometrial and ovarian cancers as Lynch syndrome–related cancers to prompt tumor testing for Lynch syndrome.[324,325]

The lifetime risk of ovarian carcinoma in females with Lynch syndrome is estimated to be as high as 12%, and the reported RR of ovarian cancer has ranged from 3.6 to 13, based on families ascertained from high-risk clinics with known or suspected Lynch syndrome.[326-331] There may be differences in ovarian cancer risk depending on the Lynch syndrome–associated pathogenic variant. In PMS2-associated Lynch syndrome, one study of 284 families was unable to identify an increased risk of ovarian cancer.[332] Another prospective registry of 3,119 Lynch syndrome–pathogenic variant carriers described the cumulative risk of ovarian cancer to range from 10% to 17% in MLH1, MSH2, and MSH6 carriers. In contrast, 0 of 67 women with a pathogenic variant in PMS2 developed ovarian cancer in 303 follow-up years.[333] Overall, there are too few cases of PMS2 pathogenic variant carriers to make definitive recommendations for ovarian cancer management. Characteristics of Lynch syndrome–associated ovarian cancers may include overrepresentation of the International Federation of Gynecology and Obstetrics stages I and II at diagnosis (reported as 81.5%), underrepresentation of serous subtypes (reported as 22.9%), and a better 10-year survival (reported as 80.6%) than reported both in population-based series and in carriers of BRCA pathogenic variants.[334,335]

The issue of breast cancer risk in Lynch syndrome has been controversial. Retrospective studies have been inconsistent, but several have demonstrated microsatellite instability in a proportion of breast cancers from individuals with Lynch syndrome;[336-339] one of these studies evaluated breast cancer risk in individuals with Lynch syndrome and found that it is not elevated.[339] However, the largest prospective study to date of 446 unaffected carriers of pathogenic variants from the Colon Cancer Family Registry [340] who were followed for up to 10 years reported an elevated SIR of 3.95 for breast cancer (95% CI, 1.59–8.13; P = .001).[340] The same group subsequently analyzed data on 764 carriers of MMR gene pathogenic variants with a prior diagnosis of colorectal cancer. Results showed that the 10-year risk of breast cancer following colorectal cancer was 2% (95% CI, 1%–4%) and that the SIR was 1.76 (95% CI, 1.07–2.59).[341] A series from the United Kingdom composed of clinically referred Lynch syndrome kindreds, with efforts to correct for ascertainment, showed a twofold increased risk of breast cancer in 157 MLH1 carriers but not in carriers of other MMR variants.[342] Results from a meta-analysis of breast cancer risk in Lynch syndrome among 15 studies with molecular tumor testing results revealed that 62 of 122 breast cancers (51%; 95% CI, 42%–60%) in MMR pathogenic variant carriers were MMR-deficient. In addition, breast cancer risk estimates among a total of 21 studies showed an increased risk of twofold to 18-fold in eight studies that compared MMR variant carriers with noncarriers, while 13 studies did not observe statistical evidence for an association of breast cancer risk with Lynch syndrome.[343]

A number of subsequent studies have suggested the presence of higher breast cancer risks than previously published,[344-347] although this has not been consistently observed.[348] Through a study of 325 Canadian families with Lynch syndrome, primarily encompassing MLH1 and MSH2 carriers, the lifetime cumulative risk for breast cancer among MSH2 carriers was reported to be 22%.[344] Similarly, breast cancer risks were elevated in a study of 423 women with Lynch syndrome, with substantially higher risks among those with MSH6 and PMS2 pathogenic variants, compared with MLH1 and MSH2 pathogenic variants.[345] In fact, breast cancer risk to age 60 years was 37.7% for PMS2, 31.1% for MSH6, 16.1% for MSH2, and 15.5% for MLH1. These findings are consistent with another study of 528 patients with Lynch syndrome–associated pathogenic variants (including MLH1, MSH2, MSH6, PMS2, and EPCAM) in which PMS2 and MSH6 variants were much more frequent among patients with only breast cancer, compared with those with only colorectal cancer (P = 2.3 x 10-5).[346] Additional data to support an association of MSH6 with breast cancer were provided through a study of over 10,000 cancer patients across the United States who had genetic testing.[347] Findings indicated that MSH6 was associated with breast cancer with an odds ratio (OR) of 2.59 (95% CI, 1.35–5.44). Taken together, these studies highlight how the risk profile among patients with Lynch syndrome is continuing to evolve as more individuals are tested through multigene panel testing, with representation of larger numbers of individuals with PMS2 and MSH6 pathogenic variants compared with prior studies. In the absence of definitive risk estimates, individuals with Lynch syndrome are screened for breast cancer on the basis of family history.[349]

Refer to the Lynch Syndrome section of the Clinical Management of Other Hereditary Breast and/or Gynecologic Cancer Syndromes section of this summary for information about clinical management of Lynch syndrome.

Li-Fraumeni syndrome (LFS)

Breast cancer is also a component of the rare LFS, in which germline variants of the TP53 gene on chromosome 17p have been documented. Located on chromosome 17p, TP53 encodes a 53kd nuclear phosphoprotein that binds DNA sequences and functions as a negative regulator of cell growth and proliferation in the setting of DNA damage. It is also an active component of programmed cell death.[350] Inactivation of the TP53 gene or disruption of the protein product is thought to allow the persistence of damaged DNA and the possible development of malignant cells.[351,352] Widely used clinical diagnostic criteria for LFS were originally developed by Chompret et al. in 2001 (called the Chompret Criteria) [353] and revised in 2009 based on additional emerging data.[354]

LFS is characterized by premenopausal breast cancer in combination with childhood sarcoma, brain tumors, leukemia, and adrenocortical carcinoma.[351,355,356]

Germline variants in TP53 are thought to account for fewer than 1% of breast cancer cases.[357] TP53-associated breast cancer is often HER2/neu-positive, in addition to being ER-positive, PR-positive, or both.[358-360] Evidence also exists that patients treated for a TP53-related tumor with chemotherapy or radiation therapy may be at risk of a treatment-related second malignancy.

Historical criteria for defining LFS

The term Li-Fraumeni syndrome was used for the first time in 1982,[361] and the following criteria, which subsequently became the classical definition of the syndrome, were proposed by Li and Fraumeni in 1988 [362]:

  1. Sarcoma before age 45 years;
  2. An FDR with cancer before age 45 years; AND
  3. Another close relative (FDR or second-degree relative [SDR]) with either cancer before age 45 years or a sarcoma at any age.

Subsequently in 2001, Chompret et al. [353] systematically developed clinical criteria for recommending TP53 genetic testing, with the narrow LFS tumor spectrum defined as sarcoma, brain tumors, breast cancer, and adrenocortical carcinoma. The criteria were as follows:

  1. A proband affected by a narrow-spectrum tumor before age 36 years AND at least one FDR or SDR affected by a narrow-spectrum tumor (other than breast cancer if the proband is affected by breast cancer) before age 46 years or multiple primary tumors; OR
  2. A proband with multiple primary tumors, two of which belong to the narrow spectrum and the first of which occurred before age 36 years, irrespective of family history; OR
  3. A proband with adrenocortical carcinoma irrespective of the age at onset and family history.

These criteria were revised in 2009 [354] based on additional emerging data [352,363] as follows:

  1. A proband with a tumor belonging to the LFS tumor spectrum* before age 46 years AND at least one FDR or SDR with an LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; OR
  2. A proband with multiple tumors (except multiple breast tumors), two of which belong to the LFS tumor spectrum and the first of which occurred before age 46 years; OR
  3. A patient with adrenocortical carcinoma or choroid plexus, irrespective of family history.

*The 2009 Chompret criteria defined the LFS tumor spectrum as including the following cancers: soft tissue sarcoma, osteosarcoma, brain tumor, premenopausal breast cancer, adrenocortical carcinoma, leukemia, and lung bronchoalveolar cancer.

In 2015, Bougeard et al. [356] revised the criteria based on data from 415 carriers of pathogenic variants, to include the presence of childhood anaplastic rhabdomyosarcoma and breast cancer before age 31 years as an indication for testing, similar to what is recommended for choroid plexus carcinoma and adrenocortical carcinoma. The criteria were revised as follows:

  1. A proband with a tumor belonging to the LFS tumor spectrum** before age 46 years AND at least one FDR or SDR with LFS tumor (except breast cancer if proband has breast cancer) before age 56 years or with multiple tumors; OR
  2. A proband with multiple tumors (except multiple breast tumors), two of which belong to the LFS tumor spectrum and the first of which occurred before age 46 years; OR
  3. A patient with adrenocortical carcinoma, choroid plexus tumor, or rhabdomyosarcoma of embryonal anaplastic subtype, irrespective of family history; OR
  4. Breast cancer before age 31 years.

**The 2015 Chompret criteria defined the LFS tumor spectrum as including the following cancers: premenopausal breast cancer, soft tissue sarcoma, osteosarcoma, central nervous system (CNS) tumor, and adrenocortical carcinoma.

Clinical characteristics of LFS

Germline TP53 pathogenic variants were identified in 17% (n = 91) of 525 samples submitted to City of Hope laboratories for clinical TP53 testing.[352] All families with a TP53 pathogenic variant had at least one family member with a sarcoma, breast cancer, brain cancer, or adrenocortical cancer (core cancers). In addition, all eight individuals with a choroid plexus tumor had a TP53 pathogenic variant, as did 14 of the 21 individuals with childhood adrenocortical cancer. In women aged 30 to 49 years who had breast cancer but no family history of other core cancers, no TP53 variants were found.

Subsequently, a large clinical series of patients from France who were tested primarily based on the 2009 version of the Chompret criteria [354] included 415 carriers of pathogenic variants from 214 families.[356] In this study, 43% of carriers had multiple malignancies, and the mean age at first tumor onset was 24.9 years. The childhood tumor spectrum was characterized by osteosarcomas, adrenocortical carcinomas, CNS tumors, and soft tissue sarcomas (present in 23%–30% collectively), whereas the adult tumor spectrum primarily encompassed breast cancer (79% of females) and soft tissue sarcomas (27% of carriers). The TP53 pathogenic variant detection rate was 6% among females younger than 31 years with breast cancer and no additional features suggestive of LFS. Evaluation of genotype-phenotype correlations indicated a gradient of clinical severity, with a significantly lower mean age at onset among those with dominant-negative missense variants (21.3 years), compared with those with all types of loss-of-function variants (28.5 years) or genomic rearrangements (35.8 years). With the exception of adrenocortical carcinoma, affected children mostly harbored dominant-negative missense pathogenic variants. Among 127 female carriers of pathogenic variants with breast cancer, 31% developed CBC. Receptor status information was available for 40 tumors, which indicated 55% were HER2-positive, and 37% were triple-positive (i.e., ER-positive, PR-positive, and HER2-positive). There was an exceptionally high rate of multiple malignancies (43%) among carriers of pathogenic variants, of which 83% were metachronous. Treatment records were available for 64 carriers who received radiation therapy for treatment of their first tumor; of these, 19 (30%) developed 26 secondary tumors within a radiation field, with a latency of 2 to 26 years (mean, 10.7 y).

Similarly, results of 286 TP53 pathogenic variant–positive individuals in the National Cancer Institute’s LFS Study indicated a cumulative cancer incidence of almost 100% by age 70 years for both males and females.[364] They reported substantial variations by sex, age, and cancer type. Specifically, cumulative cancer incidence reached 50% by age 31 years in females and age 46 years in males, although male risks were higher in childhood and late adulthood. Cumulative cancer incidence by sex for the top four cancers is included in Table 8. Of those with one cancer, 49% developed at least one additional cancer after a median of 10 years. Age-specific risks for developing first and second cancers were comparable.

Table 8. Cumulative Cancer Risks for the Most Common Li-Fraumeni Syndrome (LFS)-Associated Cancers a,b
  Cumulative Cancer Risk by Age 70 Years
aAdapted from Mai et al.[364]
bOther cancers, such as adrenocortical carcinoma, leukemia, and lung bronchoalveolar cancer, have been considered part of the LFS cancer spectrum.[354,356]
Cancer Type Females (%) Males (%)
Breast cancer 54
Soft tissue sarcoma 15 22
Brain cancer 6 19
Osteosarcoma 5 11

With the increasing use of multigene (panel) tests, it is important to recognize that pathogenic variants in TP53 are unexpectedly being identified in individuals without a family history characteristic of LFS.[365] The clinical significance of finding an isolated TP53 pathogenic variant in an individual or family who does not meet the Chompret criteria is uncertain. Consequently, it remains important to interpret cancer risks and determine optimal management strategies for individuals who are unexpectedly found to have a germline TP53 pathogenic variant, while considering their personal and family histories.

One cohort study evaluated 116 individuals with a germline TP53 pathogenic variant yearly at the National Institutes of Health Clinical Center using multimodality screening with and without gadolinium. Baseline screening identified a cancer in eight patients (6.9%) with a false-positive rate of 34.5% for MRI (n = 40).[366] Screening for breast cancer with annual breast MRI is recommended;[99] additional screening for other cancers has been studied and is evolving.[367,368]

PTEN hamartoma tumor syndromes (including Cowden syndrome)

Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome (BRRS) are part of a spectrum of conditions known collectively as PTEN hamartoma tumor syndromes (PHTS). Approximately 85% of patients diagnosed with Cowden syndrome, and approximately 60% of patients with BRRS have an identifiable PTEN pathogenic variant.[369] In addition, PTEN pathogenic variants have been identified in patients with very diverse clinical phenotypes.[370] The term PHTS refers to any patient with a PTEN pathogenic variant, irrespective of clinical presentation.

PTEN functions as a dual-specificity phosphatase that removes phosphate groups from tyrosine, serine, and threonine. PTEN pathogenic variants are diverse and can present as nonsense, missense, frameshift, or splice-site variants. Approximately 40% of variants are found in exon 5, which encodes the phosphatase core motif; several recurrent pathogenic variants have been observed at this location.[371] Pathogenic variants in the 5’ end of PTEN or within the phosphatase core of PTEN tend to affect more organ systems.[372]

Operational criteria for the diagnosis of Cowden syndrome have been published and subsequently updated.[373,374] These include major, minor, and pathognomonic criteria that consist of certain mucocutaneous manifestations and adult-onset dysplastic gangliocytoma of the cerebellum (Lhermitte-Duclos disease). An updated set of criteria based on a systematic literature review has been suggested [375] and is currently utilized in the National Comprehensive Cancer Network (NCCN) guidelines.[99] Contrary to previous criteria, the authors concluded that there was insufficient evidence for any features to be classified as pathognomonic. Increased genetic testing (especially multigene panels) has identified individuals with germline PTEN pathogenic variants who do not meet diagnostic criteria for PHTS. Diagnostic criteria will need to be reconciled with these recently discovered phenotypes. Hence, it is unclear whether PHTS diagnoses should be based on clinical features or a positive PTEN genetic test result. The American College of Medical Genetics and Genomics (ACMG) suggests that referral for genetics consultation be considered for individuals with a personal history of or a first-degree relative with the following: 1) adult-onset Lhermitte-Duclos disease or 2) any three of the major or minor criteria that have been established for the diagnosis of Cowden syndrome.[101] Detailed recommendations, including diagnostic criteria for Cowden syndrome, can be found in the NCCN and ACMG guidelines.[99,101] Additionally, a predictive model that uses clinical criteria to estimate the probability of a PTEN pathogenic variant is available; a cost-effectiveness analysis suggests that germline PTEN testing is cost effective if the probability of a variant is greater than 10%.[376]

Over a 10-year period, the International Cowden Consortium (ICC) prospectively recruited a consecutive series of adult and pediatric patients meeting relaxed ICC criteria for PTEN testing in the United States, Europe, and Asia.[377] Most individuals did not meet the clinical criteria for a diagnosis of Cowden syndrome or BRRS. Of the 3,399 individuals recruited and tested, 295 probands (8.8%) and an additional 73 family members carried a germline PTEN pathogenic variant. The authors concluded that melanoma, kidney cancer, and colorectal cancer should be added to the spectrum of cancers associated with PTEN germline pathogenic variants (in addition to breast cancer, thyroid cancer, and endometrial cancer). This conclusion was based on the high melanoma, kidney, and colorectal cancer lifetime risk estimates found in individuals with PTEN pathogenic variants. A second study of approximately 100 patients with a germline PTEN pathogenic variant confirmed these findings and suggested a cumulative cancer risk of 85% by age 70 years.[378]

Although PTEN pathogenic variants, which are estimated to occur in 1 in 200,000 individuals,[373] account for a small fraction of hereditary breast cancer, the characterization of PTEN function will provide valuable insights into the signal pathway and the maintenance of normal cell physiology.[373,379] Lifetime breast cancer risk is estimated to be between 25% and 50% among women with Cowden syndrome.[380] Other studies have reported risks as high as 85%;[377,378,381,382] however, there are concerns regarding selection bias in these studies. As in other forms of hereditary breast cancer, onset is often at a young age and may be bilateral.[383] Lifetime risk of endometrial cancer is estimated to be between 19% and 28%, depending on the cohort studied, with an increased risk of premenopausal onset.[377,378,384] Because of the low prevalence of PTEN pathogenic variants in the population, the proportion of endometrial cancer attributable to Cowden syndrome is small. There are no data that link PTEN pathogenic variants to an increased risk of ovarian cancer. Skin manifestations include multiple trichilemmomas, oral fibromas and papillomas, and acral, palmar, and plantar keratoses. History or observation of the characteristic skin features raises a suspicion of Cowden syndrome. CNS manifestations include macrocephaly, developmental delay, and dysplastic gangliocytomas of the cerebellum.[385,386] (Refer to the PDQ summaries on Genetics of Colorectal Cancer and Genetics of Skin Cancer for more information about PTEN hamartoma tumor syndromes [including Cowden syndrome].)

Diffuse gastric and lobular breast cancer syndrome

The E-cadherin gene CDH1 was first described in 1998 in three Maori families with multiple cases of diffuse gastric cancer (DGC), leading to the designation of hereditary diffuse gastric cancer (HDGC). There have been multiple subsequent reports of an excess of lobular breast cancer in HDGC families.[387] CDH1 is located on chromosome 16q22.1 and encodes the E-cadherin protein, a calcium-dependent homophilic adhesion molecule that plays a key role in cellular adhesion, cell polarity, cell signaling, and maintenance of cellular differentiation and tissue morphology.[388] E-cadherin binds to various catenins to stabilize the cytoplasmic cell adhesion complex and to maintain the E-cadherin interaction with actin filament.[389] Loss of CDH1 can occur as a result of somatic mutations, LOH, or hypermethylation, and can result in dedifferentiation and invasiveness in human cancers.[390,391] Classic histopathologic findings in gastrectomy specimens include in situ signet ring cells and/or pagetoid spread of signet ring cells. Of all gastric cancers, 1% to 3% are attributed to inherited gastric cancer syndromes.[392]

HDGC is an autosomal dominant syndrome associated with poorly differentiated invasive adenocarcinoma of the stomach presenting as linitis plastica. It is a highly penetrant and highly fatal syndrome, with a risk of clinical DGC ranging from 40% to 83%.[387] The risk of lobular breast cancer, which is characterized by small uniform cells that tend to invade in “single files,” is also increased in HDGC. Although invasive lobular breast cancer represents only 10% to 15% of all breast cancers, the lifetime risk of lobular breast cancer in carriers of CDH1 pathogenic variants ranges from 30% to 50%.[389,390] Guidelines for screening for CDH1 vary but include multiple cases of DGC in a family, early age of DGC, or lobular breast cancer in a family with DGC. Approximately 25% of families meeting these criteria are found to have a pathogenic variant in CDH1.[392] CDH1 pathogenic variants have been found in some families with lobular breast cancer but no gastric cancer.[393] The management of individuals with CDH1 pathogenic variants without a family history of gastric cancer is unclear.[393]

Peutz-Jeghers syndrome (PJS)

PJS is an early-onset autosomal dominant disorder characterized by melanocytic macules on the lips, the perioral region, and buccal region; and multiple gastrointestinal polyps, both hamartomatous and adenomatous.[394-396] Germline pathogenic variants in the STK11 gene at chromosome 19p13.3 have been identified in the vast majority of PJS families.[397-401] The most common cancers in PJS are gastrointestinal. However, other organs are at increased risk of developing malignancies. For example, the cumulative risks have been estimated to be 32% to 54% for breast cancer [402-404] and 21% for ovarian cancer (mainly ovarian sex-cord tumors).[402] The risk for pancreatic cancer has been estimated to be more than 100-fold higher than that in the general population.[402] A systematic review found a lifetime cumulative cancer risk, all sites combined, of up to 93% in patients with PJS.[402,405] Table 9 shows the cumulative risk of these tumors.

Females with PJS are also predisposed to the development of cervical adenoma malignum, a rare and very aggressive adenocarcinoma of the cervix.[406] In addition, females with PJS commonly develop benign ovarian sex-cord tumors with annular tubules, whereas males with PJS are predisposed to development of Sertoli-cell testicular tumors;[407] although neither of these two tumor types is malignant, they can cause symptoms related to increased estrogen production.

Although the risk of malignancy appears to be exceedingly high in individuals with PJS based on the published literature, the possibility that selection and referral biases have resulted in overestimates of these risks should be considered.

Table 9. Cumulative Cancer Risks in Peutz-Jeghers Syndrome Up To Specified Age a
Site Age (y) Cumulative Risk (%)b Reference(s)
GI = gastrointestinal.
aReprinted with permission from Macmillan Publishers Ltd: Gastroenterology [405], copyright 2010.
bAll cumulative risks were increased compared with the general population (P < .05), with the exception of cervix and testes.
cGI cancers include colorectal, small intestinal, gastric, esophageal, and pancreatic.
dWesterman et al.: GI cancer does not include pancreatic cancer.[408]
eDid not include adenoma malignum of the cervix or Sertoli cell tumors of the testes.
Any cancer 60–70 37–93 [401-404,408,409]
GI cancerc,d 60–70 38–66 [403,404,408,409]
Gynecological cancer 60–70 13–18 [403,404]
Per origin      
Stomach 65 29 [402]
Small bowel 65 13 [402]
Colorectum 65 39 [402,403]
Pancreas 65–70 11–36 [402,403]
Lung 65–70 7–17 [402-404]
Breast 60–70 32–54 [402-404]
Uterus 65 9 [402]
Ovary 65 21 [402]
Cervixe 65 10 [402]
Testese 65 9 [402]
Peutz-Jeghers gene(s)

PJS is caused by pathogenic variants in the STK11 (also called LKB1) tumor suppressor gene located on chromosome 19p13.[398,399] Unlike the adenomas seen in familial adenomatous polyposis, the polyps arising in PJS are hamartomas. Studies of the hamartomatous polyps and cancers of PJS show allelic imbalance (LOH) consistent with the two-hit hypothesis, demonstrating that STK11 is a tumor suppressor gene.[410,411] However, heterozygous STK11 knockout mice develop hamartomas without inactivation of the remaining wild-type allele, suggesting that haploinsufficiency may be sufficient for initial tumor development in PJS.[412] Subsequently, the cancers that develop in STK11 +/- mice do show LOH;[413] indeed, compound mutant mice heterozygous for pathogenic variants in STK11 +/- and homozygous for pathogenic variants in TP53 -/- have accelerated development of both hamartomas and cancers.[414]

Germline variants of the STK11 gene represent a spectrum of nonsense, frameshift, and missense variants, and splice-site variants and large deletions.[397,403]

Approximately 85% of variants are localized to regions of the kinase domain of the expressed protein. No strong genotype-phenotype correlations have been identified.[403] Up to 30% of variants are large deletions involving one or more exons of STK11, underscoring the importance of deletion analysis in suspected cases of PJS.[397]

STK11 has been unequivocally demonstrated to cause PJS. Although earlier estimates using direct DNA sequencing showed a 50% pathogenic variant detection rate in STK11, studies adding techniques to detect large deletions have found pathogenic variants in up to 94% of individuals meeting clinical criteria for PJS.[397,405,415] Given the results of these studies, it is unlikely that other major genes cause PJS.

Clinical management

The high cumulative risk of cancers in PJS has led to the various screening recommendations summarized in the table of Published Recommendations for Diagnosis and Surveillance of Peutz-Jeghers Syndrome (PJS) in the PDQ summary on Genetics of Colorectal Cancer.

PALB2

PALB2 (partner and localizer of BRCA2) interacts with the BRCA2 protein and plays a role in homologous recombination and double-stranded DNA repair. Similar to BRIP1 and BRCA2, biallelic pathogenic variants in PALB2 have also been shown to cause Fanconi anemia.[416]

PALB2 pathogenic variants have been screened for in multiple small studies of familial and early-onset breast cancer in multiple populations.[14,417-434] Pathogenic variant prevalence has ranged from 0.4% to 3.9%. Similar to BRIP1 and CHEK2, there was incomplete segregation of PALB2 pathogenic variants in families with hereditary breast cancer.[417] Among 559 cases with CBC and 565 matched controls with unilateral breast cancer, pathogenic (truncating) PALB2 pathogenic variants were identified in 0.9% of cases and in none of the controls (RR, 5.3; 95% CI, 1.8–13.2).[428]

Data based on 154 families with loss-of-function PALB2 variants suggest that this gene may be an important cause of hereditary breast cancer, with risks that overlap with BRCA2.[435] In this study, analysis of 362 family members from 154 families with PALB2 pathogenic variants indicated that the absolute risk of female breast cancer by age 70 years ranged from 33% (95% CI, 24%–44%) for those with no family history of breast cancer to 58% (95% CI, 50%–66%) for those with two or more FDRs with early-onset breast cancer. Furthermore, among 63 breast cancer cases in which HER2 status was known, 30% had triple-negative disease. An earlier Finnish study reported on a PALB2 founder pathogenic variant (c.1592delT) that confers a 40% risk of breast cancer to age 70 years [418] and is associated with a high incidence (54%) of triple-negative disease and lower survival.[419] Pathogenic variants have been observed in early-onset and familial breast cancer in many populations.[420,421] A large report of 1,824 patients with TNBC unselected for family history, recruited through 12 studies, identified 1.2% with a PALB2 pathogenic variant.[249] (Refer to the BRCA1 pathology section of this summary for more information about this study.)

In a later Polish study of more than 12,529 unselected women with breast cancer and 4,702 controls, PALB2 pathogenic variants were detected in 116 cases (0.93%; 95% CI, 0.76%–1.09%) and 10 controls (0.21%; 95% CI, 0.08%–0.34%), with an OR for breast cancer of 4.39 (95% CI, 2.30–8.37).[436] The study findings confirm a substantially elevated risk of breast cancer (24%–40%) among women with a PALB2 pathogenic variant up to age 75 years. The 5-year cumulative incidence of CBC was 10% among those with a PALB2 pathogenic variant, compared with 17% among those with a BRCA1 pathogenic variant and 3% among those without a variant in either gene. Furthermore, the 10-year survival for women with a PALB2 pathogenic variant and breast cancer was 48% (95% CI, 36.5%–63.2%), compared with 72.0% among those with a BRCA1 pathogenic variant and 74.7% among those without a variant in either gene. Among PALB2 carriers, breast tumors 2 cm or larger had substantially worse outcomes (32.4% 10-year survival), compared with tumors smaller than 2 cm (82.4% 10-year survival). Approximately one-third of those with a PALB2 pathogenic variant had TNBC, and the average age at breast cancer diagnosis was 53.3 years.

A similar case-control study from China enrolled 16,501 unselected patients with breast cancer and 5,890 controls. These patients were screened for PALB2, BRCA1, and BRCA2 pathogenic variants. The prevalence of PALB2 pathogenic variants was 0.97% in the cases and 0.19% in the controls. The OR for breast cancer for carriers was 5.23 (95% CI, 2.84–9.65; P > .0001). PALB2 carriers were more likely to be age 30 years or younger (6.88% vs. 3.56%; P = .04). PALB2 carriers were also more likely to have TNBCs (22.83% vs. 13.56%; P = .004), larger tumors (tumor size ≥2 cm, 55.93% vs. 45.93%; P = .04), node-positive tumors (49.60% vs. 38.80%; P = .018), and contralateral breast cancers (6.29% vs. 2.01%; P = .003). Additionally, PALB2 carriers were more likely to have a family history of breast and/or ovarian cancers (20.63% vs. 7.96%; P < .0001).[437]

In the largest study to date, 524 families with pathogenic variants in PALB2 recruited through an international effort, the RR of breast cancer in women was reported to be 7.18 (95% CI, 5.82–8.85), with a 53% risk (95% CI, 44%–63%) of breast cancer to age 80 years.[438] Additional elevated risks reported included ovarian cancer (RR, 2.91; 95% CI, 1.40–6.04) with a lifetime risk of 5% (95% CI, 2%–10%), pancreatic cancer (RR, 2.37; 95% CI, 1.24–4.50) with a lifetime risk of 2% to 3% (95% CI for women, 1%–4%; 95% CI for men, 2%–5%), and male breast cancer (RR, 7.34; 95% CI, 1.28–42.18) with a lifetime risk of 1% (95% CI, 0.2%–5%). These findings confirm the role of PALB2 as an inherited breast cancer gene in women, while firmly establishing an association with ovarian, pancreatic, and male breast cancers.

Male breast cancer has been observed in PALB2 pathogenic variant–positive breast cancer families.[14,422,435] In a study of 115 male breast cancer cases in which 18 men had BRCA2 pathogenic variants, an additional two men had either a pathogenic or predicted pathogenic PALB2 variant (accounting for about 10% of germline variants in the study and 1%–2% of the total sample).[14] The RR of breast cancer for male carriers of PALB2 pathogenic variants compared with that seen in the general population was estimated to be 8.30 (95% CI, 0.77–88.56; P = .08) in the study of 154 families.[435]

After the identification of PALB2 pathogenic variants in pancreatic tumors and the detection of germline pathogenic variants in 3% of 96 familial pancreatic patients,[439] numerous studies have pointed to a role for PALB2 in pancreatic cancer. PALB2 pathogenic variants were detected in 3.7% of 81 familial pancreatic cancer families [440] and in 2.1% of 94 BRCA1/BRCA2 pathogenic variant–negative breast cancer patients who had either a personal or family history of pancreatic cancer.[441] Two relatively small studies—one of 77 BRCA1/BRCA2 pathogenic variant–negative probands with a personal or family history of pancreatic cancer, one-half of whom were of AJ descent, and another study of 29 Italian pancreatic cancer patients with a personal or family history of breast or ovarian cancer—failed to detect any PALB2 pathogenic variants.[442,443] A sixfold increase in pancreatic cancer was observed in the relatives of 33 BRCA1/BRCA2-negative, PALB2 pathogenic variant–positive breast cancer probands.[422]

Overall, the observed prevalence of PALB2 pathogenic variants in familial breast cancer varied depending on ascertainment relative to personal and family history of pancreatic and ovarian cancers, but in all studies, the observed pathogenic variant rate was lower than 4%. Data suggest that the RR of breast cancer may overlap with that of BRCA2, particularly in those with a strong family history; thus, it remains important to refine cancer risk estimates in larger studies. Furthermore, the risk of other cancers (e.g., pancreatic) is poorly defined. Given the low PALB2 pathogenic variant prevalence in the population, additional data are needed to define best candidates for testing and appropriate management.

De Novo Pathogenic Variant Rate

Until the 1990s, the diagnosis of genetically inherited breast and ovarian cancer syndromes was based on clinical manifestations and family history. Now that some of the genes involved in these syndromes have been identified, a few studies have attempted to estimate the spontaneous pathogenic variant rate (de novo pathogenic variant rate) in these populations. Interestingly, PJS, PTEN hamartoma syndromes, and LFS are all thought to have high rates of spontaneous pathogenic variants, in the 10% to 30% range,[444-447] while estimates of de novo pathogenic variants in the BRCA genes are thought to be low, primarily on the basis of the few case reports published.[448-456] Additionally, there has been only one case series of breast cancer patients who were tested for BRCA pathogenic variants in which a de novo variant was identified. Specifically, in this study of 193 patients with sporadic breast cancer, 17 pathogenic variants were detected, one of which was confirmed to be a de novo pathogenic variant.[448] As such, the de novo pathogenic variant rate appears to be low and fall into the 5% or less range, based on the limited studies performed.[448-456] Similarly, estimates of de novo pathogenic variants in the MMR genes associated with Lynch syndrome are thought to be low, in the 0.9% to 5% range.[457-459] However, these estimates of spontaneous pathogenic variant rates in the BRCA genes and Lynch syndrome genes seem to overlap with the estimates of nonpaternity rates in various populations (0.6%–3.3%),[460-462] making the de novo pathogenic variant rate for these genes relatively low.

References
  1. Phipps RF, Perry PM: Familial breast cancer. Postgrad Med J 64 (757): 847-9, 1988. [PUBMED Abstract]
  2. Sellers TA, Potter JD, Rich SS, et al.: Familial clustering of breast and prostate cancers and risk of postmenopausal breast cancer. J Natl Cancer Inst 86 (24): 1860-5, 1994. [PUBMED Abstract]
  3. Newman B, Austin MA, Lee M, et al.: Inheritance of human breast cancer: evidence for autosomal dominant transmission in high-risk families. Proceedings of the National Academy of Sciences 85 (9): 3044-48, 1988.
  4. Hall JM, Lee MK, Newman B, et al.: Linkage of early-onset familial breast cancer to chromosome 17q21. Science 250 (4988): 1684-9, 1990. [PUBMED Abstract]
  5. Narod SA, Feunteun J, Lynch HT, et al.: Familial breast-ovarian cancer locus on chromosome 17q12-q23. Lancet 338 (8759): 82-3, 1991. [PUBMED Abstract]
  6. 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 94 (18): 1365-72, 2002. [PUBMED Abstract]
  7. Thompson D, Easton DF; Breast Cancer Linkage Consortium: Cancer Incidence in BRCA1 mutation carriers. J Natl Cancer Inst 94 (18): 1358-65, 2002. [PUBMED Abstract]
  8. Risch HA, McLaughlin JR, Cole DE, et al.: Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario, Canada. J Natl Cancer Inst 98 (23): 1694-706, 2006. [PUBMED Abstract]
  9. Tai YC, Domchek S, Parmigiani G, et al.: Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 99 (23): 1811-4, 2007. [PUBMED Abstract]
  10. Wooster R, Neuhausen SL, Mangion J, et al.: Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science 265 (5181): 2088-90, 1994. [PUBMED Abstract]
  11. Gayther SA, Mangion J, Russell P, et al.: Variation of risks of breast and ovarian cancer associated with different germline mutations of the BRCA2 gene. Nat Genet 15 (1): 103-5, 1997. [PUBMED Abstract]
  12. Cancer risks in BRCA2 mutation carriers. The Breast Cancer Linkage Consortium. J Natl Cancer Inst 91 (15): 1310-6, 1999. [PUBMED Abstract]
  13. Liede A, Karlan BY, Narod SA: Cancer risks for male carriers of germline mutations in BRCA1 or BRCA2: a review of the literature. J Clin Oncol 22 (4): 735-42, 2004. [PUBMED Abstract]
  14. Ding YC, Steele L, Kuan CJ, et al.: Mutations in BRCA2 and PALB2 in male breast cancer cases from the United States. Breast Cancer Res Treat 126 (3): 771-8, 2011. [PUBMED Abstract]
  15. Tonin P, Weber B, Offit K, et al.: Frequency of recurrent BRCA1 and BRCA2 mutations in Ashkenazi Jewish breast cancer families. Nat Med 2 (11): 1179-83, 1996. [PUBMED Abstract]
  16. Easton DF, Bishop DT, Ford D, et al.: Genetic linkage analysis in familial breast and ovarian cancer: results from 214 families. The Breast Cancer Linkage Consortium. Am J Hum Genet 52 (4): 678-701, 1993. [PUBMED Abstract]
  17. Venkitaraman AR: Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 108 (2): 171-82, 2002. [PUBMED Abstract]
  18. Narod SA, Foulkes WD: BRCA1 and BRCA2: 1994 and beyond. Nat Rev Cancer 4 (9): 665-76, 2004. [PUBMED Abstract]
  19. An Open Access On-Line Breast Cancer Mutation Data Base [Database]. Bethesda, Md: National Human Genome Research Institute, 2002. Available online. Last accessed February 4, 2022.
  20. Ford D, Easton DF, Peto J: Estimates of the gene frequency of BRCA1 and its contribution to breast and ovarian cancer incidence. Am J Hum Genet 57 (6): 1457-62, 1995. [PUBMED Abstract]
  21. Whittemore AS, Gong G, John EM, et al.: Prevalence of BRCA1 mutation carriers among U.S. non-Hispanic Whites. Cancer Epidemiol Biomarkers Prev 13 (12): 2078-83, 2004. [PUBMED Abstract]
  22. Eng C, Brody LC, Wagner TM, et al.: Interpreting epidemiological research: blinded comparison of methods used to estimate the prevalence of inherited mutations in BRCA1. J Med Genet 38 (12): 824-33, 2001. [PUBMED Abstract]
  23. Unger MA, Nathanson KL, Calzone K, et al.: Screening for genomic rearrangements in families with breast and ovarian cancer identifies BRCA1 mutations previously missed by conformation-sensitive gel electrophoresis or sequencing. Am J Hum Genet 67 (4): 841-50, 2000. [PUBMED Abstract]
  24. Walsh T, Casadei S, Coats KH, et al.: Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer. JAMA 295 (12): 1379-88, 2006. [PUBMED Abstract]
  25. Palma MD, Domchek SM, Stopfer J, et al.: The relative contribution of point mutations and genomic rearrangements in BRCA1 and BRCA2 in high-risk breast cancer families. Cancer Res 68 (17): 7006-14, 2008. [PUBMED Abstract]
  26. Stadler ZK, Saloustros E, Hansen NA, et al.: Absence of genomic BRCA1 and BRCA2 rearrangements in Ashkenazi breast and ovarian cancer families. Breast Cancer Res Treat 123 (2): 581-5, 2010. [PUBMED Abstract]
  27. Judkins T, Rosenthal E, Arnell C, et al.: Clinical significance of large rearrangements in BRCA1 and BRCA2. Cancer 118 (21): 5210-6, 2012. [PUBMED Abstract]
  28. Shannon KM, Rodgers LH, Chan-Smutko G, et al.: Which individuals undergoing BRACAnalysis need BART testing? Cancer Genet 204 (8): 416-22, 2011. [PUBMED Abstract]
  29. Jackson SA, Davis AA, Li J, et al.: Characteristics of individuals with breast cancer rearrangements in BRCA1 and BRCA2. Cancer 120 (10): 1557-64, 2014. [PUBMED Abstract]
  30. Weitzel JN, Lagos VI, Herzog JS, et al.: Evidence for common ancestral origin of a recurring BRCA1 genomic rearrangement identified in high-risk Hispanic families. Cancer Epidemiol Biomarkers Prev 16 (8): 1615-20, 2007. [PUBMED Abstract]
  31. Plon SE, Eccles DM, Easton D, et al.: Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results. Hum Mutat 29 (11): 1282-91, 2008. [PUBMED Abstract]
  32. 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 20 (6): 1480-90, 2002. [PUBMED Abstract]
  33. 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 294 (15): 1925-33, 2005. [PUBMED Abstract]
  34. Hall MJ, Reid JE, Burbidge LA, et al.: BRCA1 and BRCA2 mutations in women of different ethnicities undergoing testing for hereditary breast-ovarian cancer. Cancer 115 (10): 2222-33, 2009. [PUBMED Abstract]
  35. Szabo C, Masiello A, Ryan JF, et al.: The breast cancer information core: database design, structure, and scope. Hum Mutat 16 (2): 123-31, 2000. [PUBMED Abstract]
  36. Spurdle AB, Healey S, Devereau A, et al.: ENIGMA--evidence-based network for the interpretation of germline mutant alleles: an international initiative to evaluate risk and clinical significance associated with sequence variation in BRCA1 and BRCA2 genes. Hum Mutat 33 (1): 2-7, 2012. [PUBMED Abstract]
  37. Goldgar DE, Easton DF, Deffenbaugh AM, et al.: Integrated evaluation of DNA sequence variants of unknown clinical significance: application to BRCA1 and BRCA2. Am J Hum Genet 75 (4): 535-44, 2004. [PUBMED Abstract]
  38. Thompson D, Easton DF, Goldgar DE: A full-likelihood method for the evaluation of causality of sequence variants from family data. Am J Hum Genet 73 (3): 652-5, 2003. [PUBMED Abstract]
  39. Spearman AD, Sweet K, Zhou XP, et al.: Clinically applicable models to characterize BRCA1 and BRCA2 variants of uncertain significance. J Clin Oncol 26 (33): 5393-400, 2008. [PUBMED Abstract]
  40. Gómez García EB, Oosterwijk JC, Timmermans M, et al.: A method to assess the clinical significance of unclassified variants in the BRCA1 and BRCA2 genes based on cancer family history. Breast Cancer Res 11 (1): R8, 2009. [PUBMED Abstract]
  41. Goldgar DE, Easton DF, Byrnes GB, et al.: Genetic evidence and integration of various data sources for classifying uncertain variants into a single model. Hum Mutat 29 (11): 1265-72, 2008. [PUBMED Abstract]
  42. Fleming MA, Potter JD, Ramirez CJ, et al.: Understanding missense mutations in the BRCA1 gene: an evolutionary approach. Proc Natl Acad Sci U S A 100 (3): 1151-6, 2003. [PUBMED Abstract]
  43. Tavtigian SV, Deffenbaugh AM, Yin L, et al.: Comprehensive statistical study of 452 BRCA1 missense substitutions with classification of eight recurrent substitutions as neutral. J Med Genet 43 (4): 295-305, 2006. [PUBMED Abstract]
  44. Mirkovic N, Marti-Renom MA, Weber BL, et al.: Structure-based assessment of missense mutations in human BRCA1: implications for breast and ovarian cancer predisposition. Cancer Res 64 (11): 3790-7, 2004. [PUBMED Abstract]
  45. Abkevich V, Zharkikh A, Deffenbaugh AM, et al.: Analysis of missense variation in human BRCA1 in the context of interspecific sequence variation. J Med Genet 41 (7): 492-507, 2004. [PUBMED Abstract]
  46. Couch FJ, Rasmussen LJ, Hofstra R, et al.: Assessment of functional effects of unclassified genetic variants. Hum Mutat 29 (11): 1314-26, 2008. [PUBMED Abstract]
  47. Chenevix-Trench G, Healey S, Lakhani S, et al.: Genetic and histopathologic evaluation of BRCA1 and BRCA2 DNA sequence variants of unknown clinical significance. Cancer Res 66 (4): 2019-27, 2006. [PUBMED Abstract]
  48. Ostrow KL, McGuire V, Whittemore AS, et al.: The effects of BRCA1 missense variants V1804D and M1628T on transcriptional activity. Cancer Genet Cytogenet 153 (2): 177-80, 2004. [PUBMED Abstract]
  49. Wu K, Hinson SR, Ohashi A, et al.: Functional evaluation and cancer risk assessment of BRCA2 unclassified variants. Cancer Res 65 (2): 417-26, 2005. [PUBMED Abstract]
  50. Weitzel JN, Lagos V, Blazer KR, et al.: Prevalence of BRCA mutations and founder effect in high-risk Hispanic families. Cancer Epidemiol Biomarkers Prev 14 (7): 1666-71, 2005. [PUBMED Abstract]
  51. Mefford HC, Baumbach L, Panguluri RC, et al.: Evidence for a BRCA1 founder mutation in families of West African ancestry. Am J Hum Genet 65 (2): 575-8, 1999. [PUBMED Abstract]
  52. 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 149 (1): 223-7, 2015. [PUBMED Abstract]
  53. Frey MK, Kopparam RV, Ni Zhou Z, et al.: Prevalence of nonfounder BRCA1/2 mutations in Ashkenazi Jewish patients presenting for genetic testing at a hereditary breast and ovarian cancer center. Cancer 125 (5): 690-697, 2019. [PUBMED Abstract]
  54. Prevalence and penetrance of BRCA1 and BRCA2 mutations in a population-based series of breast cancer cases. Anglian Breast Cancer Study Group. Br J Cancer 83 (10): 1301-8, 2000. [PUBMED Abstract]
  55. Papelard H, de Bock GH, van Eijk R, et al.: Prevalence of BRCA1 in a hospital-based population of Dutch breast cancer patients. Br J Cancer 83 (6): 719-24, 2000. [PUBMED Abstract]
  56. Loman N, Johannsson O, Kristoffersson U, et al.: Family history of breast and ovarian cancers and BRCA1 and BRCA2 mutations in a population-based series of early-onset breast cancer. J Natl Cancer Inst 93 (16): 1215-23, 2001. [PUBMED Abstract]
  57. 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 66 (16): 8297-308, 2006. [PUBMED Abstract]
  58. Newman B, Mu H, Butler LM, et al.: Frequency of breast cancer attributable to BRCA1 in a population-based series of American women. JAMA 279 (12): 915-21, 1998. [PUBMED Abstract]
  59. Basham VM, Lipscombe JM, Ward JM, et al.: BRCA1 and BRCA2 mutations in a population-based study of male breast cancer. Breast Cancer Res 4 (1): R2, 2002. [PUBMED Abstract]
  60. Risch HA, McLaughlin JR, Cole DE, et al.: Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a population series of 649 women with ovarian cancer. Am J Hum Genet 68 (3): 700-10, 2001. [PUBMED Abstract]
  61. Rubin SC, Blackwood MA, Bandera C, et al.: BRCA1, BRCA2, and hereditary nonpolyposis colorectal cancer gene mutations in an unselected ovarian cancer population: relationship to family history and implications for genetic testing. Am J Obstet Gynecol 178 (4): 670-7, 1998. [PUBMED Abstract]
  62. Pal T, Permuth-Wey J, Betts JA, et al.: BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases. Cancer 104 (12): 2807-16, 2005. [PUBMED Abstract]
  63. Struewing JP, Hartge P, Wacholder S, et al.: The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336 (20): 1401-8, 1997. [PUBMED Abstract]
  64. Gabai-Kapara E, Lahad A, Kaufman B, et al.: Population-based screening for breast and ovarian cancer risk due to BRCA1 and BRCA2. Proc Natl Acad Sci U S A 111 (39): 14205-10, 2014. [PUBMED Abstract]
  65. Metcalfe KA, Poll A, Royer R, et al.: Screening for founder mutations in BRCA1 and BRCA2 in unselected Jewish women. J Clin Oncol 28 (3): 387-91, 2010. [PUBMED Abstract]
  66. King MC, Marks JH, Mandell JB, et al.: Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science 302 (5645): 643-6, 2003. [PUBMED Abstract]
  67. 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 36 (8): 983-6, 2000. [PUBMED Abstract]
  68. 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 36 (5): 369-73, 1999. [PUBMED Abstract]
  69. Struewing JP, Coriaty ZM, Ron E, et al.: Founder BRCA1/2 mutations among male patients with breast cancer in Israel. Am J Hum Genet 65 (6): 1800-2, 1999. [PUBMED Abstract]
  70. Hirsh-Yechezkel G, Chetrit A, Lubin F, et al.: Population attributes affecting the prevalence of BRCA mutation carriers in epithelial ovarian cancer cases in Israel. Gynecol Oncol 89 (3): 494-8, 2003. [PUBMED Abstract]
  71. 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 66 (4): 1259-72, 2000. [PUBMED Abstract]
  72. Levine DA, Argenta PA, Yee CJ, et al.: Fallopian tube and primary peritoneal carcinomas associated with BRCA mutations. J Clin Oncol 21 (22): 4222-7, 2003. [PUBMED Abstract]
  73. John EM, Miron A, Gong G, et al.: Prevalence of pathogenic BRCA1 mutation carriers in 5 US racial/ethnic groups. JAMA 298 (24): 2869-76, 2007. [PUBMED Abstract]
  74. 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 31 (2): 210-6, 2013. [PUBMED Abstract]
  75. Torres-Mejía G, Royer R, Llacuachaqui M, et al.: Recurrent BRCA1 and BRCA2 mutations in Mexican women with breast cancer. Cancer Epidemiol Biomarkers Prev 24 (3): 498-505, 2015. [PUBMED Abstract]
  76. 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 118 (3): 299-302, 2010. [PUBMED Abstract]
  77. Aziz S, Kuperstein G, Rosen B, et al.: A genetic epidemiological study of carcinoma of the fallopian tube. Gynecol Oncol 80 (3): 341-5, 2001. [PUBMED Abstract]
  78. Manchanda R, Loggenberg K, Sanderson S, et al.: Population testing for cancer predisposing BRCA1/BRCA2 mutations in the Ashkenazi-Jewish community: a randomized controlled trial. J Natl Cancer Inst 107 (1): 379, 2015. [PUBMED Abstract]
  79. Manchanda R, Legood R, Burnell M, et al.: Cost-effectiveness of population screening for BRCA mutations in Ashkenazi jewish women compared with family history-based testing. J Natl Cancer Inst 107 (1): 380, 2015. [PUBMED Abstract]
  80. Metcalfe KA, Poll A, Royer R, et al.: A comparison of the detection of BRCA mutation carriers through the provision of Jewish population-based genetic testing compared with clinic-based genetic testing. Br J Cancer 109 (3): 777-9, 2013. [PUBMED Abstract]
  81. Manchanda R, Patel S, Antoniou AC, et al.: Cost-effectiveness of population based BRCA testing with varying Ashkenazi Jewish ancestry. Am J Obstet Gynecol 217 (5): 578.e1-578.e12, 2017. [PUBMED Abstract]
  82. Manchanda R, Patel S, Gordeev VS, et al.: Cost-effectiveness of Population-Based BRCA1, BRCA2, RAD51C, RAD51D, BRIP1, PALB2 Mutation Testing in Unselected General Population Women. J Natl Cancer Inst 110 (7): 714-725, 2018. [PUBMED Abstract]
  83. Couch FJ, DeShano ML, Blackwood MA, et al.: BRCA1 mutations in women attending clinics that evaluate the risk of breast cancer. N Engl J Med 336 (20): 1409-15, 1997. [PUBMED Abstract]
  84. Shattuck-Eidens D, Oliphant A, McClure M, et al.: BRCA1 sequence analysis in women at high risk for susceptibility mutations. Risk factor analysis and implications for genetic testing. JAMA 278 (15): 1242-50, 1997. [PUBMED Abstract]
  85. Spiegelman D, Colditz GA, Hunter D, et al.: Validation of the Gail et al. model for predicting individual breast cancer risk. J Natl Cancer Inst 86 (8): 600-7, 1994. [PUBMED Abstract]
  86. Frank TS, Manley SA, Olopade OI, et al.: Sequence analysis of BRCA1 and BRCA2: correlation of mutations with family history and ovarian cancer risk. J Clin Oncol 16 (7): 2417-25, 1998. [PUBMED Abstract]
  87. Chang-Claude J, Dong J, Schmidt S, et al.: Using gene carrier probability to select high risk families for identifying germline mutations in breast cancer susceptibility genes. J Med Genet 35 (2): 116-21, 1998. [PUBMED Abstract]
  88. Couch FJ, Hartmann LC: BRCA1 testing--advances and retreats. JAMA 279 (12): 955-7, 1998. [PUBMED Abstract]
  89. Parmigiani G, Berry D, Aguilar O: Determining carrier probabilities for breast cancer-susceptibility genes BRCA1 and BRCA2. Am J Hum Genet 62 (1): 145-58, 1998. [PUBMED Abstract]
  90. Ready K, Litton JK, Arun BK: Clinical application of breast cancer risk assessment models. Future Oncol 6 (3): 355-65, 2010. [PUBMED Abstract]
  91. Amir E, Freedman OC, Seruga B, et al.: Assessing women at high risk of breast cancer: a review of risk assessment models. J Natl Cancer Inst 102 (10): 680-91, 2010. [PUBMED Abstract]
  92. Lakhani SR, Reis-Filho JS, Fulford L, et al.: Prediction of BRCA1 status in patients with breast cancer using estrogen receptor and basal phenotype. Clin Cancer Res 11 (14): 5175-80, 2005. [PUBMED Abstract]
  93. Kwon JS, Gutierrez-Barrera AM, Young D, et al.: Expanding the criteria for BRCA mutation testing in breast cancer survivors. J Clin Oncol 28 (27): 4214-20, 2010. [PUBMED Abstract]
  94. Young SR, Pilarski RT, Donenberg T, et al.: The prevalence of BRCA1 mutations among young women with triple-negative breast cancer. BMC Cancer 9: 86, 2009. [PUBMED Abstract]
  95. Greenup R, Buchanan A, Lorizio W, et al.: Prevalence of BRCA mutations among women with triple-negative breast cancer (TNBC) in a genetic counseling cohort. Ann Surg Oncol 20 (10): 3254-8, 2013. [PUBMED Abstract]
  96. Fasching PA, Yadav S, Hu C, et al.: Mutations in BRCA1/2 and Other Panel Genes in Patients With Metastatic Breast Cancer -Association With Patient and Disease Characteristics and Effect on Prognosis. J Clin Oncol 39 (15): 1619-1630, 2021. [PUBMED Abstract]
  97. Stuttgen K, Croessmann S, Fetting J, et al.: Pathogenic Germline Variants in Patients With Metastatic Breast Cancer. JAMA Oncol 5 (10): 1506-1508, 2019. [PUBMED Abstract]
  98. Robson ME, Storm CD, Weitzel J, et al.: American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 28 (5): 893-901, 2010. [PUBMED Abstract]
  99. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2022. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2022. Available online with free registration. Last accessed March 16, 2022.
  100. Statement of the American Society of Human Genetics on genetic testing for breast and ovarian cancer predisposition. Am J Hum Genet 55 (5): i-iv, 1994. [PUBMED Abstract]
  101. Hampel H, Bennett RL, Buchanan A, et al.: A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med 17 (1): 70-87, 2015. [PUBMED Abstract]
  102. Owens DK, Davidson KW, Krist AH, et al.: Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 322 (7): 652-665, 2019. [PUBMED Abstract]
  103. Lancaster JM, Powell CB, Kauff ND, et al.: Society of Gynecologic Oncologists Education Committee statement on risk assessment for inherited gynecologic cancer predispositions. Gynecol Oncol 107 (2): 159-62, 2007. [PUBMED Abstract]
  104. 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 41 (6): 474-80, 2004. [PUBMED Abstract]
  105. Apicella C, Dowty JG, Dite GS, et al.: Validation study of the LAMBDA model for predicting the BRCA1 or BRCA2 mutation carrier status of North American Ashkenazi Jewish women. Clin Genet 72 (2): 87-97, 2007. [PUBMED Abstract]
  106. Antoniou AC, Pharoah PP, Smith P, et al.: The BOADICEA model of genetic susceptibility to breast and ovarian cancer. Br J Cancer 91 (8): 1580-90, 2004. [PUBMED Abstract]
  107. Struewing JP, Abeliovich D, Peretz T, et al.: The carrier frequency of the BRCA1 185delAG mutation is approximately 1 percent in Ashkenazi Jewish individuals. Nat Genet 11 (2): 198-200, 1995. [PUBMED Abstract]
  108. Oddoux C, Struewing JP, Clayton CM, et al.: The carrier frequency of the BRCA2 6174delT mutation among Ashkenazi Jewish individuals is approximately 1%. Nat Genet 14 (2): 188-90, 1996. [PUBMED Abstract]
  109. Warner E, Foulkes W, Goodwin P, et al.: Prevalence and penetrance of BRCA1 and BRCA2 gene mutations in unselected Ashkenazi Jewish women with breast cancer. J Natl Cancer Inst 91 (14): 1241-7, 1999. [PUBMED Abstract]
  110. Euhus DM, Smith KC, Robinson L, et al.: Pretest prediction of BRCA1 or BRCA2 mutation by risk counselors and the computer model BRCAPRO. J Natl Cancer Inst 94 (11): 844-51, 2002. [PUBMED Abstract]
  111. de la Hoya M, Díez O, Pérez-Segura P, et al.: Pre-test prediction models of BRCA1 or BRCA2 mutation in breast/ovarian families attending familial cancer clinics. J Med Genet 40 (7): 503-10, 2003. [PUBMED Abstract]
  112. Katki HA: Incorporating medical interventions into carrier probability estimation for genetic counseling. BMC Med Genet 8: 13, 2007. [PUBMED Abstract]
  113. Weitzel JN, Lagos VI, Cullinane CA, et al.: Limited family structure and BRCA gene mutation status in single cases of breast cancer. JAMA 297 (23): 2587-95, 2007. [PUBMED Abstract]
  114. Jervis S, Song H, Lee A, et al.: A risk prediction algorithm for ovarian cancer incorporating BRCA1, BRCA2, common alleles and other familial effects. J Med Genet 52 (7): 465-75, 2015. [PUBMED Abstract]
  115. Oros KK, Ghadirian P, Maugard CM, et al.: Application of BRCA1 and BRCA2 mutation carrier prediction models in breast and/or ovarian cancer families of French Canadian descent. Clin Genet 70 (4): 320-9, 2006. [PUBMED Abstract]
  116. Vogel KJ, Atchley DP, Erlichman J, et al.: BRCA1 and BRCA2 genetic testing in Hispanic patients: mutation prevalence and evaluation of the BRCAPRO risk assessment model. J Clin Oncol 25 (29): 4635-41, 2007. [PUBMED Abstract]
  117. Kurian AW, Gong GD, John EM, et al.: Performance of prediction models for BRCA mutation carriage in three racial/ethnic groups: findings from the Northern California Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev 18 (4): 1084-91, 2009. [PUBMED Abstract]
  118. Kurian AW, Gong GD, Chun NM, et al.: Performance of BRCA1/2 mutation prediction models in Asian Americans. J Clin Oncol 26 (29): 4752-8, 2008. [PUBMED Abstract]
  119. Berry DA, Parmigiani G, Sanchez J, et al.: Probability of carrying a mutation of breast-ovarian cancer gene BRCA1 based on family history. J Natl Cancer Inst 89 (3): 227-38, 1997. [PUBMED Abstract]
  120. Barcenas CH, Hosain GM, Arun B, et al.: Assessing BRCA carrier probabilities in extended families. J Clin Oncol 24 (3): 354-60, 2006. [PUBMED Abstract]
  121. Kang HH, Williams R, Leary J, et al.: Evaluation of models to predict BRCA germline mutations. Br J Cancer 95 (7): 914-20, 2006. [PUBMED Abstract]
  122. 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 45 (7): 425-31, 2008. [PUBMED Abstract]
  123. Fischer C, Kuchenbäcker K, Engel C, et al.: Evaluating the performance of the breast cancer genetic risk models BOADICEA, IBIS, BRCAPRO and Claus for predicting BRCA1/2 mutation carrier probabilities: a study based on 7352 families from the German Hereditary Breast and Ovarian Cancer Consortium. J Med Genet 50 (6): 360-7, 2013. [PUBMED Abstract]
  124. Biswas S, Tankhiwale N, Blackford A, et al.: Assessing the added value of breast tumor markers in genetic risk prediction model BRCAPRO. Breast Cancer Res Treat 133 (1): 347-55, 2012. [PUBMED Abstract]
  125. Tai YC, Chen S, Parmigiani G, et al.: Incorporating tumor immunohistochemical markers in BRCA1 and BRCA2 carrier prediction. Breast Cancer Res 10 (2): 401, 2008. [PUBMED Abstract]
  126. Tyrer J, Duffy SW, Cuzick J: A breast cancer prediction model incorporating familial and personal risk factors. Stat Med 23 (7): 1111-30, 2004. [PUBMED Abstract]
  127. Ready KJ, Vogel KJ, Atchley DP, et al.: Accuracy of the BRCAPRO model among women with bilateral breast cancer. Cancer 115 (4): 725-30, 2009. [PUBMED Abstract]
  128. Huo D, Senie RT, Daly M, et al.: Prediction of BRCA Mutations Using the BRCAPRO Model in Clinic-Based African American, Hispanic, and Other Minority Families in the United States. J Clin Oncol 27 (8): 1184-90, 2009. [PUBMED Abstract]
  129. Daniels MS, Babb SA, King RH, et al.: Underestimation of risk of a BRCA1 or BRCA2 mutation in women with high-grade serous ovarian cancer by BRCAPRO: a multi-institution study. J Clin Oncol 32 (12): 1249-55, 2014. [PUBMED Abstract]
  130. 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 72 (5): 1117-30, 2003. [PUBMED Abstract]
  131. Chen S, Parmigiani G: Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol 25 (11): 1329-33, 2007. [PUBMED Abstract]
  132. Kuchenbaecker KB, Hopper JL, Barnes DR, et al.: Risks of Breast, Ovarian, and Contralateral Breast Cancer for BRCA1 and BRCA2 Mutation Carriers. JAMA 317 (23): 2402-2416, 2017. [PUBMED Abstract]
  133. Antoniou AC, Pharoah PD, Narod S, et al.: Breast and ovarian cancer risks to carriers of the BRCA1 5382insC and 185delAG and BRCA2 6174delT mutations: a combined analysis of 22 population based studies. J Med Genet 42 (7): 602-3, 2005. [PUBMED Abstract]
  134. Finkelman BS, Rubinstein WS, Friedman S, et al.: Breast and ovarian cancer risk and risk reduction in Jewish BRCA1/2 mutation carriers. J Clin Oncol 30 (12): 1321-8, 2012. [PUBMED Abstract]
  135. Ramus SJ, Song H, Dicks E, et al.: Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer. J Natl Cancer Inst 107 (11): , 2015. [PUBMED Abstract]
  136. Song H, Dicks E, Ramus SJ, et al.: Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population. J Clin Oncol 33 (26): 2901-7, 2015. [PUBMED Abstract]
  137. Rebbeck TR, Mitra N, Wan F, et al.: Association of type and location of BRCA1 and BRCA2 mutations with risk of breast and ovarian cancer. JAMA 313 (13): 1347-61, 2015. [PUBMED Abstract]
  138. Rebbeck TR, Friebel TM, Mitra N, et al.: Inheritance of deleterious mutations at both BRCA1 and BRCA2 in an international sample of 32,295 women. Breast Cancer Res 18 (1): 112, 2016. [PUBMED Abstract]
  139. Guindalini RS, Song A, Fackenthal JD, et al.: Genetic anticipation in BRCA1/BRCA2 families after controlling for ascertainment bias and cohort effect. Cancer 122 (12): 1913-20, 2016. [PUBMED Abstract]
  140. Agranat S, Baris H, Kedar I, et al.: Earlier Age of Breast Cancer Onset in Israeli BRCA Carriers-Is it a Real Phenomenon? Breast J 22 (6): 662-666, 2016. [PUBMED Abstract]
  141. Litton JK, Ready K, Chen H, et al.: Earlier age of onset of BRCA mutation-related cancers in subsequent generations. Cancer 118 (2): 321-5, 2012. [PUBMED Abstract]
  142. Wang WW, Spurdle AB, Kolachana P, et al.: A single nucleotide polymorphism in the 5' untranslated region of RAD51 and risk of cancer among BRCA1/2 mutation carriers. Cancer Epidemiol Biomarkers Prev 10 (9): 955-60, 2001. [PUBMED Abstract]
  143. Levy-Lahad E, Lahad A, Eisenberg S, et al.: A single nucleotide polymorphism in the RAD51 gene modifies cancer risk in BRCA2 but not BRCA1 carriers. Proc Natl Acad Sci U S A 98 (6): 3232-6, 2001. [PUBMED Abstract]
  144. Narod SA: Modifiers of risk of hereditary breast and ovarian cancer. Nat Rev Cancer 2 (2): 113-23, 2002. [PUBMED Abstract]
  145. Kramer JL, Velazquez IA, Chen BE, et al.: Prophylactic oophorectomy reduces breast cancer penetrance during prospective, long-term follow-up of BRCA1 mutation carriers. J Clin Oncol 23 (34): 8629-35, 2005. [PUBMED Abstract]
  146. Antoniou AC, Spurdle AB, Sinilnikova OM, et al.: Common breast cancer-predisposition alleles are associated with breast cancer risk in BRCA1 and BRCA2 mutation carriers. Am J Hum Genet 82 (4): 937-48, 2008. [PUBMED Abstract]
  147. 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 46 (12): 2275-84, 2010. [PUBMED Abstract]
  148. Antoniou AC, Rookus M, Andrieu N, et al.: Reproductive and hormonal factors, and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: results from the International BRCA1/2 Carrier Cohort Study. Cancer Epidemiol Biomarkers Prev 18 (2): 601-10, 2009. [PUBMED Abstract]
  149. Milne RL, Osorio A, Ramón y Cajal T, et al.: Parity and the risk of breast and ovarian cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 119 (1): 221-32, 2010. [PUBMED Abstract]
  150. Friedman E, Kotsopoulos J, Lubinski J, et al.: Spontaneous and therapeutic abortions and the risk of breast cancer among BRCA mutation carriers. Breast Cancer Res 8 (2): R15, 2006. [PUBMED Abstract]
  151. Jernström H, Lubinski J, Lynch HT, et al.: Breast-feeding and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 96 (14): 1094-8, 2004. [PUBMED Abstract]
  152. Breast Cancer Family Registry, Kathleen Cuningham Consortium for Research into Familial Breast Cancer (Australasia), Ontario Cancer Genetics Network (Canada): Smoking and risk of breast cancer in carriers of mutations in BRCA1 or BRCA2 aged less than 50 years. Breast Cancer Res Treat 109 (1): 67-75, 2008. [PUBMED Abstract]
  153. Antoniou AC, Beesley J, McGuffog L, et al.: Common breast cancer susceptibility alleles and the risk of breast cancer for BRCA1 and BRCA2 mutation carriers: implications for risk prediction. Cancer Res 70 (23): 9742-54, 2010. [PUBMED Abstract]
  154. Ramus SJ, Kartsonaki C, Gayther SA, et al.: Genetic variation at 9p22.2 and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 103 (2): 105-16, 2011. [PUBMED Abstract]
  155. Milne RL, Antoniou AC: Genetic modifiers of cancer risk for BRCA1 and BRCA2 mutation carriers. Ann Oncol 22 (Suppl 1): i11-7, 2011. [PUBMED Abstract]
  156. Graeser MK, Engel C, Rhiem K, et al.: Contralateral breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Clin Oncol 27 (35): 5887-92, 2009. [PUBMED Abstract]
  157. Malone KE, Begg CB, Haile RW, et al.: Population-based study of the risk of second primary contralateral breast cancer associated with carrying a mutation in BRCA1 or BRCA2. J Clin Oncol 28 (14): 2404-10, 2010. [PUBMED Abstract]
  158. 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 124 (3): 643-51, 2010. [PUBMED Abstract]
  159. Metcalfe K, Gershman S, Lynch HT, et al.: Predictors of contralateral breast cancer in BRCA1 and BRCA2 mutation carriers. Br J Cancer 104 (9): 1384-92, 2011. [PUBMED Abstract]
  160. Molina-Montes E, Pérez-Nevot B, Pollán M, et al.: Cumulative risk of second primary contralateral breast cancer in BRCA1/BRCA2 mutation carriers with a first breast cancer: A systematic review and meta-analysis. Breast 23 (6): 721-42, 2014. [PUBMED Abstract]
  161. Basu NN, Ingham S, Hodson J, et al.: Risk of contralateral breast cancer in BRCA1 and BRCA2 mutation carriers: a 30-year semi-prospective analysis. Fam Cancer 14 (4): 531-8, 2015. [PUBMED Abstract]
  162. van den Broek AJ, van 't Veer LJ, Hooning MJ, et al.: Impact of Age at Primary Breast Cancer on Contralateral Breast Cancer Risk in BRCA1/2 Mutation Carriers. J Clin Oncol 34 (5): 409-18, 2016. [PUBMED Abstract]
  163. Pierce LJ, Phillips KA, Griffith KA, et al.: Local therapy in BRCA1 and BRCA2 mutation carriers with operable breast cancer: comparison of breast conservation and mastectomy. Breast Cancer Res Treat 121 (2): 389-98, 2010. [PUBMED Abstract]
  164. Reiner AS, Robson ME, Mellemkjær L, et al.: Radiation Treatment, ATM, BRCA1/2, and CHEK2*1100delC Pathogenic Variants and Risk of Contralateral Breast Cancer. J Natl Cancer Inst 112 (12): 1275-1279, 2020. [PUBMED Abstract]
  165. Domchek SM, Jhaveri K, Patil S, et al.: Risk of metachronous breast cancer after BRCA mutation-associated ovarian cancer. Cancer 119 (7): 1344-8, 2013. [PUBMED Abstract]
  166. Vencken PM, Kriege M, Hooning M, et al.: The risk of primary and contralateral breast cancer after ovarian cancer in BRCA1/BRCA2 mutation carriers: Implications for counseling. Cancer 119 (5): 955-62, 2013. [PUBMED Abstract]
  167. Gangi A, Cass I, Paik D, et al.: Breast cancer following ovarian cancer in BRCA mutation carriers. JAMA Surg 149 (12): 1306-13, 2014. [PUBMED Abstract]
  168. McGee J, Giannakeas V, Karlan B, et al.: Risk of breast cancer after a diagnosis of ovarian cancer in BRCA mutation carriers: Is preventive mastectomy warranted? Gynecol Oncol 145 (2): 346-351, 2017. [PUBMED Abstract]
  169. 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 97 (2): 457-67, 2005. [PUBMED Abstract]
  170. 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 296 (2): 185-92, 2006. [PUBMED Abstract]
  171. Evans DG, Susnerwala I, Dawson J, et al.: Risk of breast cancer in male BRCA2 carriers. J Med Genet 47 (10): 710-1, 2010. [PUBMED Abstract]
  172. Edwards SM, Kote-Jarai Z, Meitz J, et al.: Two percent of men with early-onset prostate cancer harbor germline mutations in the BRCA2 gene. Am J Hum Genet 72 (1): 1-12, 2003. [PUBMED Abstract]
  173. Giusti RM, Rutter JL, Duray PH, et al.: A twofold increase in BRCA mutation related prostate cancer among Ashkenazi Israelis is not associated with distinctive histopathology. J Med Genet 40 (10): 787-92, 2003. [PUBMED Abstract]
  174. van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, et al.: Cancer risks in BRCA2 families: estimates for sites other than breast and ovary. J Med Genet 42 (9): 711-9, 2005. [PUBMED Abstract]
  175. Mersch J, Jackson MA, Park M, et al.: Cancers associated with BRCA1 and BRCA2 mutations other than breast and ovarian. Cancer 121 (2): 269-75, 2015. [PUBMED Abstract]
  176. 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 98 (2): 502-7, 2008. [PUBMED Abstract]
  177. Tryggvadóttir L, Vidarsdóttir L, Thorgeirsson T, et al.: Prostate cancer progression and survival in BRCA2 mutation carriers. J Natl Cancer Inst 99 (12): 929-35, 2007. [PUBMED Abstract]
  178. Agalliu I, Gern R, Leanza S, et al.: Associations of high-grade prostate cancer with BRCA1 and BRCA2 founder mutations. Clin Cancer Res 15 (3): 1112-20, 2009. [PUBMED Abstract]
  179. Narod SA, Neuhausen S, Vichodez G, et al.: Rapid progression of prostate cancer in men with a BRCA2 mutation. Br J Cancer 99 (2): 371-4, 2008. [PUBMED Abstract]
  180. Edwards SM, Evans DG, Hope Q, et al.: Prostate cancer in BRCA2 germline mutation carriers is associated with poorer prognosis. Br J Cancer 103 (6): 918-24, 2010. [PUBMED Abstract]
  181. Gallagher DJ, Gaudet MM, Pal P, et al.: Germline BRCA mutations denote a clinicopathologic subset of prostate cancer. Clin Cancer Res 16 (7): 2115-21, 2010. [PUBMED Abstract]
  182. Couch FJ, Johnson MR, Rabe KG, et al.: The prevalence of BRCA2 mutations in familial pancreatic cancer. Cancer Epidemiol Biomarkers Prev 16 (2): 342-6, 2007. [PUBMED Abstract]
  183. Hahn SA, Greenhalf B, Ellis I, et al.: BRCA2 germline mutations in familial pancreatic carcinoma. J Natl Cancer Inst 95 (3): 214-21, 2003. [PUBMED Abstract]
  184. Murphy KM, Brune KA, Griffin C, et al.: Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, and BRCA2 in familial pancreatic cancer: deleterious BRCA2 mutations in 17%. Cancer Res 62 (13): 3789-93, 2002. [PUBMED Abstract]
  185. Real FX, Malats N, Lesca G, et al.: Family history of cancer and germline BRCA2 mutations in sporadic exocrine pancreatic cancer. Gut 50 (5): 653-7, 2002. [PUBMED Abstract]
  186. Dagan E: Predominant Ashkenazi BRCA1/2 mutations in families with pancreatic cancer. Genet Test 12 (2): 267-71, 2008. [PUBMED Abstract]
  187. Goggins M, Schutte M, Lu J, et al.: Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Res 56 (23): 5360-4, 1996. [PUBMED Abstract]
  188. Lal G, Liu G, Schmocker B, et al.: Inherited predisposition to pancreatic adenocarcinoma: role of family history and germ-line p16, BRCA1, and BRCA2 mutations. Cancer Res 60 (2): 409-16, 2000. [PUBMED Abstract]
  189. Ozçelik H, Schmocker B, Di Nicola N, et al.: Germline BRCA2 6174delT mutations in Ashkenazi Jewish pancreatic cancer patients. Nat Genet 16 (1): 17-8, 1997. [PUBMED Abstract]
  190. Ferrone CR, Levine DA, Tang LH, et al.: BRCA germline mutations in Jewish patients with pancreatic adenocarcinoma. J Clin Oncol 27 (3): 433-8, 2009. [PUBMED Abstract]
  191. National Cancer Institute: DevCan: Probability of Developing or Dying of Cancer Software. Version 6.7.7. Bethesda, Md: Division of Cancer Control & Population Sciences, National Cancer Institute, 2017. Available online with free registration. Last accessed February 4, 2022.
  192. Ford D, Easton DF, Bishop DT, et al.: Risks of cancer in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Lancet 343 (8899): 692-5, 1994. [PUBMED Abstract]
  193. Anton-Culver H, Cohen PF, Gildea ME, et al.: Characteristics of BRCA1 mutations in a population-based case series of breast and ovarian cancer. Eur J Cancer 36 (10): 1200-8, 2000. [PUBMED Abstract]
  194. Peelen T, de Leeuw W, van Lent K, et al.: Genetic analysis of a breast-ovarian cancer family, with 7 cases of colorectal cancer linked to BRCA1, fails to support a role for BRCA1 in colorectal tumorigenesis. Int J Cancer 88 (5): 778-82, 2000. [PUBMED Abstract]
  195. Berman DB, Costalas J, Schultz DC, et al.: A common mutation in BRCA2 that predisposes to a variety of cancers is found in both Jewish Ashkenazi and non-Jewish individuals. Cancer Res 56 (15): 3409-14, 1996. [PUBMED Abstract]
  196. Aretini P, D'Andrea E, Pasini B, et al.: Different expressivity of BRCA1 and BRCA2: analysis of 179 Italian pedigrees with identified mutation. Breast Cancer Res Treat 81 (1): 71-9, 2003. [PUBMED Abstract]
  197. Kirchhoff T, Satagopan JM, Kauff ND, et al.: Frequency of BRCA1 and BRCA2 mutations in unselected Ashkenazi Jewish patients with colorectal cancer. J Natl Cancer Inst 96 (1): 68-70, 2004. [PUBMED Abstract]
  198. Niell BL, Rennert G, Bonner JD, et al.: BRCA1 and BRCA2 founder mutations and the risk of colorectal cancer. J Natl Cancer Inst 96 (1): 15-21, 2004. [PUBMED Abstract]
  199. Chen-Shtoyerman R, Figer A, Fidder HH, et al.: The frequency of the predominant Jewish mutations in BRCA1 and BRCA2 in unselected Ashkenazi colorectal cancer patients. Br J Cancer 84 (4): 475-7, 2001. [PUBMED Abstract]
  200. Levine DA, Lin O, Barakat RR, et al.: Risk of endometrial carcinoma associated with BRCA mutation. Gynecol Oncol 80 (3): 395-8, 2001. [PUBMED Abstract]
  201. Goshen R, Chu W, Elit L, et al.: Is uterine papillary serous adenocarcinoma a manifestation of the hereditary breast-ovarian cancer syndrome? Gynecol Oncol 79 (3): 477-81, 2000. [PUBMED Abstract]
  202. Smith A, Moran A, Boyd MC, et al.: Phenocopies in BRCA1 and BRCA2 families: evidence for modifier genes and implications for screening. J Med Genet 44 (1): 10-15, 2007. [PUBMED Abstract]
  203. Goldgar D, Venne V, Conner T, et al.: BRCA phenocopies or ascertainment bias? J Med Genet 44 (8): e86; author reply e88, 2007. [PUBMED Abstract]
  204. Eisinger F: Phenocopies: actual risk or self-fulfilling prophecy? J Med Genet 44 (8): e87; author reply e88, 2007. [PUBMED Abstract]
  205. Sasieni P: Phenocopies in families seen by cancer geneticists. J Med Genet 44 (6): e82, 2007. [PUBMED Abstract]
  206. Tilanus-Linthorst MM: No screening yet after a negative test for the family mutation. J Med Genet 44 (5): e79, 2007. [PUBMED Abstract]
  207. Katki HA, Gail MH, Greene MH: Breast-cancer risk in BRCA-mutation-negative women from BRCA-mutation-positive families. Lancet Oncol 8 (12): 1042-3, 2007. [PUBMED Abstract]
  208. Raskin L, Lejbkowicz F, Barnett-Griness O, et al.: BRCA1 breast cancer risk is modified by CYP19 polymorphisms in Ashkenazi Jews. Cancer Epidemiol Biomarkers Prev 18 (5): 1617-23, 2009. [PUBMED Abstract]
  209. Gronwald J, Cybulski C, Lubinski J, et al.: Phenocopies in breast cancer 1 (BRCA1) families: implications for genetic counselling. J Med Genet 44 (4): e76, 2007. [PUBMED Abstract]
  210. Rowan E, Poll A, Narod SA: A prospective study of breast cancer risk in relatives of BRCA1/BRCA2 mutation carriers. J Med Genet 44 (8): e89; author reply e88, 2007. [PUBMED Abstract]
  211. Vos JR, de Bock GH, Teixeira N, et al.: Proven non-carriers in BRCA families have an earlier age of onset of breast cancer. Eur J Cancer 49 (9): 2101-6, 2013. [PUBMED Abstract]
  212. Domchek SM, Gaudet MM, Stopfer JE, et al.: Breast cancer risks in individuals testing negative for a known family mutation in BRCA1 or BRCA2. Breast Cancer Res Treat 119 (2): 409-14, 2010. [PUBMED Abstract]
  213. Korde LA, Mueller CM, Loud JT, et al.: No evidence of excess breast cancer risk among mutation-negative women from BRCA mutation-positive families. Breast Cancer Res Treat 125 (1): 169-73, 2011. [PUBMED Abstract]
  214. Kurian AW, Gong GD, John EM, et al.: Breast cancer risk for noncarriers of family-specific BRCA1 and BRCA2 mutations: findings from the Breast Cancer Family Registry. J Clin Oncol 29 (34): 4505-9, 2011. [PUBMED Abstract]
  215. Harvey SL, Milne RL, McLachlan SA, et al.: Prospective study of breast cancer risk for mutation negative women from BRCA1 or BRCA2 mutation positive families. Breast Cancer Res Treat 130 (3): 1057-61, 2011. [PUBMED Abstract]
  216. Kauff ND, Mitra N, Robson ME, et al.: Risk of ovarian cancer in BRCA1 and BRCA2 mutation-negative hereditary breast cancer families. J Natl Cancer Inst 97 (18): 1382-4, 2005. [PUBMED Abstract]
  217. Lee JS, John EM, McGuire V, et al.: Breast and ovarian cancer in relatives of cancer patients, with and without BRCA mutations. Cancer Epidemiol Biomarkers Prev 15 (2): 359-63, 2006. [PUBMED Abstract]
  218. Metcalfe KA, Finch A, Poll A, et al.: Breast cancer risks in women with a family history of breast or ovarian cancer who have tested negative for a BRCA1 or BRCA2 mutation. Br J Cancer 100 (2): 421-5, 2009. [PUBMED Abstract]
  219. Liede A, Karlan BY, Baldwin RL, et al.: Cancer incidence in a population of Jewish women at risk of ovarian cancer. J Clin Oncol 20 (6): 1570-7, 2002. [PUBMED Abstract]
  220. Ingham SL, Warwick J, Buchan I, et al.: Ovarian cancer among 8,005 women from a breast cancer family history clinic: no increased risk of invasive ovarian cancer in families testing negative for BRCA1 and BRCA2. J Med Genet 50 (6): 368-72, 2013. [PUBMED Abstract]
  221. Barnes DR, Antoniou AC: Unravelling modifiers of breast and ovarian cancer risk for BRCA1 and BRCA2 mutation carriers: update on genetic modifiers. J Intern Med 271 (4): 331-43, 2012. [PUBMED Abstract]
  222. Chenevix-Trench G, Milne RL, Antoniou AC, et al.: An international initiative to identify genetic modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: the Consortium of Investigators of Modifiers of BRCA1 and BRCA2 (CIMBA). Breast Cancer Res 9 (2): 104, 2007. [PUBMED Abstract]
  223. 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 62 (3): 676-89, 1998. [PUBMED Abstract]
  224. Thompson D, Easton D; Breast Cancer Linkage Consortium: Variation in cancer risks, by mutation position, in BRCA2 mutation carriers. Am J Hum Genet 68 (2): 410-9, 2001. [PUBMED Abstract]
  225. Thompson D, Easton D; Breast Cancer Linkage Consortium: Variation in BRCA1 cancer risks by mutation position. Cancer Epidemiol Biomarkers Prev 11 (4): 329-36, 2002. [PUBMED Abstract]
  226. Scott CL, Jenkins MA, Southey MC, et al.: Average age-specific cumulative risk of breast cancer according to type and site of germline mutations in BRCA1 and BRCA2 estimated from multiple-case breast cancer families attending Australian family cancer clinics. Hum Genet 112 (5-6): 542-51, 2003. [PUBMED Abstract]
  227. Lubinski J, Phelan CM, Ghadirian P, et al.: Cancer variation associated with the position of the mutation in the BRCA2 gene. Fam Cancer 3 (1): 1-10, 2004. [PUBMED Abstract]
  228. Satagopan JM, Boyd J, Kauff ND, et al.: Ovarian cancer risk in Ashkenazi Jewish carriers of BRCA1 and BRCA2 mutations. Clin Cancer Res 8 (12): 3776-81, 2002. [PUBMED Abstract]
  229. Rennert G, Dishon S, Rennert HS, et al.: Differences in the characteristics of families with BRCA1 and BRCA2 mutations in Israel. Eur J Cancer Prev 14 (4): 357-61, 2005. [PUBMED Abstract]
  230. James PA, Sawyer S, Boyle S, et al.: Large genomic rearrangements in the familial breast and ovarian cancer gene BRCA1 are associated with an increased frequency of high risk features. Fam Cancer 14 (2): 287-95, 2015. [PUBMED Abstract]
  231. Eisinger F, Jacquemier J, Charpin C, et al.: Mutations at BRCA1: the medullary breast carcinoma revisited. Cancer Res 58 (8): 1588-92, 1998. [PUBMED Abstract]
  232. Pathology of familial breast cancer: differences between breast cancers in carriers of BRCA1 or BRCA2 mutations and sporadic cases. Breast Cancer Linkage Consortium. Lancet 349 (9064): 1505-10, 1997. [PUBMED Abstract]
  233. Armes JE, Egan AJ, Southey MC, et al.: The histologic phenotypes of breast carcinoma occurring before age 40 years in women with and without BRCA1 or BRCA2 germline mutations: a population-based study. Cancer 83 (11): 2335-45, 1998. [PUBMED Abstract]
  234. Foulkes WD, Stefansson IM, Chappuis PO, et al.: Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst 95 (19): 1482-5, 2003. [PUBMED Abstract]
  235. Verhoog LC, Brekelmans CT, Seynaeve C, et al.: Survival and tumour characteristics of breast-cancer patients with germline mutations of BRCA1. Lancet 351 (9099): 316-21, 1998. [PUBMED Abstract]
  236. Manié E, Vincent-Salomon A, Lehmann-Che J, et al.: High frequency of TP53 mutation in BRCA1 and sporadic basal-like carcinomas but not in BRCA1 luminal breast tumors. Cancer Res 69 (2): 663-71, 2009. [PUBMED Abstract]
  237. Southey MC, Ramus SJ, Dowty JG, et al.: Morphological predictors of BRCA1 germline mutations in young women with breast cancer. Br J Cancer 104 (6): 903-9, 2011. [PUBMED Abstract]
  238. Mavaddat N, Barrowdale D, Andrulis IL, et al.: Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiol Biomarkers Prev 21 (1): 134-47, 2012. [PUBMED Abstract]
  239. 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 20 (9): 2310-8, 2002. [PUBMED Abstract]
  240. Sorlie T, Tibshirani R, Parker J, et al.: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100 (14): 8418-23, 2003. [PUBMED Abstract]
  241. Lee E, McKean-Cowdin R, Ma H, et al.: Characteristics of triple-negative breast cancer in patients with a BRCA1 mutation: results from a population-based study of young women. J Clin Oncol 29 (33): 4373-80, 2011. [PUBMED Abstract]
  242. Anders C, Carey LA: Understanding and treating triple-negative breast cancer. Oncology (Williston Park) 22 (11): 1233-9; discussion 1239-40, 1243, 2008. [PUBMED Abstract]
  243. 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 26 (26): 4282-8, 2008. [PUBMED Abstract]
  244. Tung N, Wang Y, Collins LC, et al.: Estrogen receptor positive breast cancers in BRCA1 mutation carriers: clinical risk factors and pathologic features. Breast Cancer Res 12 (1): R12, 2010. [PUBMED Abstract]
  245. Lakhani SR, Khanna KK, Chenevix-Trench G: Are estrogen receptor-positive breast cancers in BRCA1 mutation carriers sporadic? Breast Cancer Res 12 (2): 104, 2010. [PUBMED Abstract]
  246. Foulkes WD, Metcalfe K, Sun P, et al.: Estrogen receptor status in BRCA1- and BRCA2-related breast cancer: the influence of age, grade, and histological type. Clin Cancer Res 10 (6): 2029-34, 2004. [PUBMED Abstract]
  247. Gonzalez-Angulo AM, Timms KM, Liu S, et al.: Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 17 (5): 1082-9, 2011. [PUBMED Abstract]
  248. Rummel S, Varner E, Shriver CD, et al.: Evaluation of BRCA1 mutations in an unselected patient population with triple-negative breast cancer. Breast Cancer Res Treat 137 (1): 119-25, 2013. [PUBMED Abstract]
  249. Couch FJ, Hart SN, Sharma P, et al.: Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer. J Clin Oncol 33 (4): 304-11, 2015. [PUBMED Abstract]
  250. Robertson L, Hanson H, Seal S, et al.: BRCA1 testing should be offered to individuals with triple-negative breast cancer diagnosed below 50 years. Br J Cancer 106 (6): 1234-8, 2012. [PUBMED Abstract]
  251. 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 85 (1): 43-8, 2014. [PUBMED Abstract]
  252. Lee LJ, Alexander B, Schnitt SJ, et al.: Clinical outcome of triple negative breast cancer in BRCA1 mutation carriers and noncarriers. Cancer 117 (14): 3093-100, 2011. [PUBMED Abstract]
  253. Eccles DM, Li N, Handwerker R, et al.: Genetic testing in a cohort of young patients with HER2-amplified breast cancer. Ann Oncol 27 (3): 467-73, 2016. [PUBMED Abstract]
  254. Foulkes WD: BRCA1 functions as a breast stem cell regulator. J Med Genet 41 (1): 1-5, 2004. [PUBMED Abstract]
  255. Perou CM, Sørlie T, Eisen MB, et al.: Molecular portraits of human breast tumours. Nature 406 (6797): 747-52, 2000. [PUBMED Abstract]
  256. Sørlie T, Perou CM, Tibshirani R, et al.: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98 (19): 10869-74, 2001. [PUBMED Abstract]
  257. Hedenfalk I, Duggan D, Chen Y, et al.: Gene-expression profiles in hereditary breast cancer. N Engl J Med 344 (8): 539-48, 2001. [PUBMED Abstract]
  258. Wessels LF, van Welsem T, Hart AA, et al.: Molecular classification of breast carcinomas by comparative genomic hybridization: a specific somatic genetic profile for BRCA1 tumors. Cancer Res 62 (23): 7110-7, 2002. [PUBMED Abstract]
  259. Palacios J, Honrado E, Osorio A, et al.: Immunohistochemical characteristics defined by tissue microarray of hereditary breast cancer not attributable to BRCA1 or BRCA2 mutations: differences from breast carcinomas arising in BRCA1 and BRCA2 mutation carriers. Clin Cancer Res 9 (10 Pt 1): 3606-14, 2003. [PUBMED Abstract]
  260. Nielsen TO, Hsu FD, Jensen K, et al.: Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10 (16): 5367-74, 2004. [PUBMED Abstract]
  261. Palacios J, Honrado E, Osorio A, et al.: Phenotypic characterization of BRCA1 and BRCA2 tumors based in a tissue microarray study with 37 immunohistochemical markers. Breast Cancer Res Treat 90 (1): 5-14, 2005. [PUBMED Abstract]
  262. Laakso M, Loman N, Borg A, et al.: Cytokeratin 5/14-positive breast cancer: true basal phenotype confined to BRCA1 tumors. Mod Pathol 18 (10): 1321-8, 2005. [PUBMED Abstract]
  263. Cheang MC, Voduc D, Bajdik C, et al.: Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res 14 (5): 1368-76, 2008. [PUBMED Abstract]
  264. Hwang ES, McLennan JL, Moore DH, et al.: Ductal carcinoma in situ in BRCA mutation carriers. J Clin Oncol 25 (6): 642-7, 2007. [PUBMED Abstract]
  265. Adem C, Reynolds C, Soderberg CL, et al.: Pathologic characteristics of breast parenchyma in patients with hereditary breast carcinoma, including BRCA1 and BRCA2 mutation carriers. Cancer 97 (1): 1-11, 2003. [PUBMED Abstract]
  266. Claus EB, Petruzella S, Matloff E, et al.: Prevalence of BRCA1 and BRCA2 mutations in women diagnosed with ductal carcinoma in situ. JAMA 293 (8): 964-9, 2005. [PUBMED Abstract]
  267. Arun B, Vogel KJ, Lopez A, et al.: High prevalence of preinvasive lesions adjacent to BRCA1/2-associated breast cancers. Cancer Prev Res (Phila Pa) 2 (2): 122-7, 2009. [PUBMED Abstract]
  268. Garber JE: BRCA1/2-associated and sporadic breast cancers: fellow travelers or not? Cancer Prev Res (Phila Pa) 2 (2): 100-3, 2009. [PUBMED Abstract]
  269. Smith KL, Adank M, Kauff N, et al.: BRCA mutations in women with ductal carcinoma in situ. Clin Cancer Res 13 (14): 4306-10, 2007. [PUBMED Abstract]
  270. Hoogerbrugge N, Bult P, Bonenkamp JJ, et al.: Numerous high-risk epithelial lesions in familial breast cancer. Eur J Cancer 42 (15): 2492-8, 2006. [PUBMED Abstract]
  271. Kauff ND, Brogi E, Scheuer L, et al.: Epithelial lesions in prophylactic mastectomy specimens from women with BRCA mutations. Cancer 97 (7): 1601-8, 2003. [PUBMED Abstract]
  272. 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) 3 (12): 1579-85, 2010. [PUBMED Abstract]
  273. Marcus JN, Watson P, Page DL, et al.: Hereditary breast cancer: pathobiology, prognosis, and BRCA1 and BRCA2 gene linkage. Cancer 77 (4): 697-709, 1996. [PUBMED Abstract]
  274. Marcus JN, Watson P, Page DL, et al.: BRCA2 hereditary breast cancer pathophenotype. Breast Cancer Res Treat 44 (3): 275-7, 1997. [PUBMED Abstract]
  275. Agnarsson BA, Jonasson JG, Björnsdottir IB, et al.: Inherited BRCA2 mutation associated with high grade breast cancer. Breast Cancer Res Treat 47 (2): 121-7, 1998. [PUBMED Abstract]
  276. Lakhani SR, Jacquemier J, Sloane JP, et al.: Multifactorial analysis of differences between sporadic breast cancers and cancers involving BRCA1 and BRCA2 mutations. J Natl Cancer Inst 90 (15): 1138-45, 1998. [PUBMED Abstract]
  277. Futreal PA, Liu Q, Shattuck-Eidens D, et al.: BRCA1 mutations in primary breast and ovarian carcinomas. Science 266 (5182): 120-2, 1994. [PUBMED Abstract]
  278. Lancaster JM, Wooster R, Mangion J, et al.: BRCA2 mutations in primary breast and ovarian cancers. Nat Genet 13 (2): 238-40, 1996. [PUBMED Abstract]
  279. Miki Y, Katagiri T, Kasumi F, et al.: Mutation analysis in the BRCA2 gene in primary breast cancers. Nat Genet 13 (2): 245-7, 1996. [PUBMED Abstract]
  280. Teng DH, Bogden R, Mitchell J, et al.: Low incidence of BRCA2 mutations in breast carcinoma and other cancers. Nat Genet 13 (2): 241-4, 1996. [PUBMED Abstract]
  281. Berchuck A, Heron KA, Carney ME, et al.: Frequency of germline and somatic BRCA1 mutations in ovarian cancer. Clin Cancer Res 4 (10): 2433-7, 1998. [PUBMED Abstract]
  282. Thompson ME, Jensen RA, Obermiller PS, et al.: Decreased expression of BRCA1 accelerates growth and is often present during sporadic breast cancer progression. Nat Genet 9 (4): 444-50, 1995. [PUBMED Abstract]
  283. Dobrovic A, Simpfendorfer D: Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res 57 (16): 3347-50, 1997. [PUBMED Abstract]
  284. Birgisdottir V, Stefansson OA, Bodvarsdottir SK, et al.: Epigenetic silencing and deletion of the BRCA1 gene in sporadic breast cancer. Breast Cancer Res 8 (4): R38, 2006. [PUBMED Abstract]
  285. Turner NC, Reis-Filho JS, Russell AM, et al.: BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 26 (14): 2126-32, 2007. [PUBMED Abstract]
  286. Rakha EA, El-Sheikh SE, Kandil MA, et al.: Expression of BRCA1 protein in breast cancer and its prognostic significance. Hum Pathol 39 (6): 857-65, 2008. [PUBMED Abstract]
  287. Wong EM, Southey MC, Fox SB, et al.: Constitutional methylation of the BRCA1 promoter is specifically associated with BRCA1 mutation-associated pathology in early-onset breast cancer. Cancer Prev Res (Phila) 4 (1): 23-33, 2011. [PUBMED Abstract]
  288. Cleton-Jansen AM, Collins N, Lakhani SR, et al.: Loss of heterozygosity in sporadic breast tumours at the BRCA2 locus on chromosome 13q12-q13. Br J Cancer 72 (5): 1241-4, 1995. [PUBMED Abstract]
  289. Hamann U, Herbold C, Costa S, et al.: Allelic imbalance on chromosome 13q: evidence for the involvement of BRCA2 and RB1 in sporadic breast cancer. Cancer Res 56 (9): 1988-90, 1996. [PUBMED Abstract]
  290. Farmer H, McCabe N, Lord CJ, et al.: Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434 (7035): 917-21, 2005. [PUBMED Abstract]
  291. Lakhani SR, Manek S, Penault-Llorca F, et al.: Pathology of ovarian cancers in BRCA1 and BRCA2 carriers. Clin Cancer Res 10 (7): 2473-81, 2004. [PUBMED Abstract]
  292. Evans DG, Young K, Bulman M, et al.: Probability of BRCA1/2 mutation varies with ovarian histology: results from screening 442 ovarian cancer families. Clin Genet 73 (4): 338-45, 2008. [PUBMED Abstract]
  293. Tonin PN, Maugard CM, Perret C, et al.: A review of histopathological subtypes of ovarian cancer in BRCA-related French Canadian cancer families. Fam Cancer 6 (4): 491-7, 2007. [PUBMED Abstract]
  294. Bolton KL, Chenevix-Trench G, Goh C, et al.: Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA 307 (4): 382-90, 2012. [PUBMED Abstract]
  295. Liu J, Cristea MC, Frankel P, et al.: Clinical characteristics and outcomes of BRCA-associated ovarian cancer: genotype and survival. Cancer Genet 205 (1-2): 34-41, 2012 Jan-Feb. [PUBMED Abstract]
  296. Crum CP, Drapkin R, Kindelberger D, et al.: Lessons from BRCA: the tubal fimbria emerges as an origin for pelvic serous cancer. Clin Med Res 5 (1): 35-44, 2007. [PUBMED Abstract]
  297. Piek JM, van Diest PJ, Zweemer RP, et al.: Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer. J Pathol 195 (4): 451-6, 2001. [PUBMED Abstract]
  298. Carcangiu ML, Radice P, Manoukian S, et al.: Atypical epithelial proliferation in fallopian tubes in prophylactic salpingo-oophorectomy specimens from BRCA1 and BRCA2 germline mutation carriers. Int J Gynecol Pathol 23 (1): 35-40, 2004. [PUBMED Abstract]
  299. Mehra K, Mehrad M, Ning G, et al.: STICS, SCOUTs and p53 signatures; a new language for pelvic serous carcinogenesis. Front Biosci (Elite Ed) 3: 625-34, 2011. [PUBMED Abstract]
  300. Powell CB, Chen LM, McLennan J, et al.: Risk-reducing salpingo-oophorectomy (RRSO) in BRCA mutation carriers: experience with a consecutive series of 111 patients using a standardized surgical-pathological protocol. Int J Gynecol Cancer 21 (5): 846-51, 2011. [PUBMED Abstract]
  301. Finch A, Shaw P, Rosen B, et al.: Clinical and pathologic findings of prophylactic salpingo-oophorectomies in 159 BRCA1 and BRCA2 carriers. Gynecol Oncol 100 (1): 58-64, 2006. [PUBMED Abstract]
  302. Medeiros F, Muto MG, Lee Y, et al.: The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome. Am J Surg Pathol 30 (2): 230-6, 2006. [PUBMED Abstract]
  303. Prat J; FIGO Committee on Gynecologic Oncology: Staging classification for cancer of the ovary, fallopian tube, and peritoneum. Int J Gynaecol Obstet 124 (1): 1-5, 2014. [PUBMED Abstract]
  304. Schorge JO, Muto MG, Lee SJ, et al.: BRCA1-related papillary serous carcinoma of the peritoneum has a unique molecular pathogenesis. Cancer Res 60 (5): 1361-4, 2000. [PUBMED Abstract]
  305. Jazaeri AA, Yee CJ, Sotiriou C, et al.: Gene expression profiles of BRCA1-linked, BRCA2-linked, and sporadic ovarian cancers. J Natl Cancer Inst 94 (13): 990-1000, 2002. [PUBMED Abstract]
  306. Gourley C, Michie CO, Roxburgh P, et al.: Increased incidence of visceral metastases in scottish patients with BRCA1/2-defective ovarian cancer: an extension of the ovarian BRCAness phenotype. J Clin Oncol 28 (15): 2505-11, 2010. [PUBMED Abstract]
  307. Vineyard MA, Daniels MS, Urbauer DL, et al.: Is low-grade serous ovarian cancer part of the tumor spectrum of hereditary breast and ovarian cancer? Gynecol Oncol 120 (2): 229-32, 2011. [PUBMED Abstract]
  308. Norquist BM, Harrell MI, Brady MF, et al.: Inherited Mutations in Women With Ovarian Carcinoma. JAMA Oncol 2 (4): 482-90, 2016. [PUBMED Abstract]
  309. Hilton JL, Geisler JP, Rathe JA, et al.: Inactivation of BRCA1 and BRCA2 in ovarian cancer. J Natl Cancer Inst 94 (18): 1396-406, 2002. [PUBMED Abstract]
  310. Quinn JE, James CR, Stewart GE, et al.: BRCA1 mRNA expression levels predict for overall survival in ovarian cancer after chemotherapy. Clin Cancer Res 13 (24): 7413-20, 2007. [PUBMED Abstract]
  311. Geisler JP, Hatterman-Zogg MA, Rathe JA, et al.: Frequency of BRCA1 dysfunction in ovarian cancer. J Natl Cancer Inst 94 (1): 61-7, 2002. [PUBMED Abstract]
  312. Vasen HF: Clinical description of the Lynch syndrome [hereditary nonpolyposis colorectal cancer (HNPCC)]. Fam Cancer 4 (3): 219-25, 2005. [PUBMED Abstract]
  313. Jascur T, Boland CR: Structure and function of the components of the human DNA mismatch repair system. Int J Cancer 119 (9): 2030-5, 2006. [PUBMED Abstract]
  314. Papadopoulos N, Nicolaides NC, Wei YF, et al.: Mutation of a mutL homolog in hereditary colon cancer. Science 263 (5153): 1625-9, 1994. [PUBMED Abstract]
  315. Peltomäki P, Vasen HF: Mutations predisposing to hereditary nonpolyposis colorectal cancer: database and results of a collaborative study. The International Collaborative Group on Hereditary Nonpolyposis Colorectal Cancer. Gastroenterology 113 (4): 1146-58, 1997. [PUBMED Abstract]
  316. Akiyama Y, Sato H, Yamada T, et al.: Germ-line mutation of the hMSH6/GTBP gene in an atypical hereditary nonpolyposis colorectal cancer kindred. Cancer Res 57 (18): 3920-3, 1997. [PUBMED Abstract]
  317. Miyaki M, Konishi M, Tanaka K, et al.: Germline mutation of MSH6 as the cause of hereditary nonpolyposis colorectal cancer. Nat Genet 17 (3): 271-2, 1997. [PUBMED Abstract]
  318. Huang J, Kuismanen SA, Liu T, et al.: MSH6 and MSH3 are rarely involved in genetic predisposition to nonpolypotic colon cancer. Cancer Res 61 (4): 1619-23, 2001. [PUBMED Abstract]
  319. Nicolaides NC, Papadopoulos N, Liu B, et al.: Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature 371 (6492): 75-80, 1994. [PUBMED Abstract]
  320. Hendriks YM, Jagmohan-Changur S, van der Klift HM, et al.: Heterozygous mutations in PMS2 cause hereditary nonpolyposis colorectal carcinoma (Lynch syndrome). Gastroenterology 130 (2): 312-22, 2006. [PUBMED Abstract]
  321. Worthley DL, Walsh MD, Barker M, et al.: Familial mutations in PMS2 can cause autosomal dominant hereditary nonpolyposis colorectal cancer. Gastroenterology 128 (5): 1431-6, 2005. [PUBMED Abstract]
  322. Vasen HF, Mecklin JP, Khan PM, et al.: The International Collaborative Group on Hereditary Non-Polyposis Colorectal Cancer (ICG-HNPCC). Dis Colon Rectum 34 (5): 424-5, 1991. [PUBMED Abstract]
  323. Vasen HF, Watson P, Mecklin JP, et al.: New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology 116 (6): 1453-6, 1999. [PUBMED Abstract]
  324. Rodriguez-Bigas MA, Boland CR, Hamilton SR, et al.: A National Cancer Institute Workshop on Hereditary Nonpolyposis Colorectal Cancer Syndrome: meeting highlights and Bethesda guidelines. J Natl Cancer Inst 89 (23): 1758-62, 1997. [PUBMED Abstract]
  325. Umar A, Boland CR, Terdiman JP, et al.: Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 96 (4): 261-8, 2004. [PUBMED Abstract]
  326. Watson P, Lynch HT: Cancer risk in mismatch repair gene mutation carriers. Fam Cancer 1 (1): 57-60, 2001. [PUBMED Abstract]
  327. Vasen HF, Wijnen JT, Menko FH, et al.: Cancer risk in families with hereditary nonpolyposis colorectal cancer diagnosed by mutation analysis. Gastroenterology 110 (4): 1020-7, 1996. [PUBMED Abstract]
  328. Aarnio M, Mecklin JP, Aaltonen LA, et al.: Life-time risk of different cancers in hereditary non-polyposis colorectal cancer (HNPCC) syndrome. Int J Cancer 64 (6): 430-3, 1995. [PUBMED Abstract]
  329. Watson P, Lynch HT: Extracolonic cancer in hereditary nonpolyposis colorectal cancer. Cancer 71 (3): 677-85, 1993. [PUBMED Abstract]
  330. Brown GJ, St John DJ, Macrae FA, et al.: Cancer risk in young women at risk of hereditary nonpolyposis colorectal cancer: implications for gynecologic surveillance. Gynecol Oncol 80 (3): 346-9, 2001. [PUBMED Abstract]
  331. Aarnio M, Sankila R, Pukkala E, et al.: Cancer risk in mutation carriers of DNA-mismatch-repair genes. Int J Cancer 81 (2): 214-8, 1999. [PUBMED Abstract]
  332. Ten Broeke SW, van der Klift HM, Tops CMJ, et al.: Cancer Risks for PMS2-Associated Lynch Syndrome. J Clin Oncol 36 (29): 2961-2968, 2018. [PUBMED Abstract]
  333. Møller P, Seppälä TT, Bernstein I, et al.: Cancer risk and survival in path_MMR carriers by gene and gender up to 75 years of age: a report from the Prospective Lynch Syndrome Database. Gut 67 (7): 1306-1316, 2018. [PUBMED Abstract]
  334. Grindedal EM, Renkonen-Sinisalo L, Vasen H, et al.: Survival in women with MMR mutations and ovarian cancer: a multicentre study in Lynch syndrome kindreds. J Med Genet 47 (2): 99-102, 2010. [PUBMED Abstract]
  335. Pal T, Permuth-Wey J, Sellers TA: A review of the clinical relevance of mismatch-repair deficiency in ovarian cancer. Cancer 113 (4): 733-42, 2008. [PUBMED Abstract]
  336. Jensen UB, Sunde L, Timshel S, et al.: Mismatch repair defective breast cancer in the hereditary nonpolyposis colorectal cancer syndrome. Breast Cancer Res Treat 120 (3): 777-82, 2010. [PUBMED Abstract]
  337. Shanley S, Fung C, Milliken J, et al.: Breast cancer immunohistochemistry can be useful in triage of some HNPCC families. Fam Cancer 8 (3): 251-5, 2009. [PUBMED Abstract]
  338. Walsh MD, Buchanan DD, Cummings MC, et al.: Lynch syndrome-associated breast cancers: clinicopathologic characteristics of a case series from the colon cancer family registry. Clin Cancer Res 16 (7): 2214-24, 2010. [PUBMED Abstract]
  339. Buerki N, Gautier L, Kovac M, et al.: Evidence for breast cancer as an integral part of Lynch syndrome. Genes Chromosomes Cancer 51 (1): 83-91, 2012. [PUBMED Abstract]
  340. Win AK, Young JP, Lindor NM, et al.: Colorectal and other cancer risks for carriers and noncarriers from families with a DNA mismatch repair gene mutation: a prospective cohort study. J Clin Oncol 30 (9): 958-64, 2012. [PUBMED Abstract]
  341. Win AK, Lindor NM, Young JP, et al.: Risks of primary extracolonic cancers following colorectal cancer in lynch syndrome. J Natl Cancer Inst 104 (18): 1363-72, 2012. [PUBMED Abstract]
  342. Harkness EF, Barrow E, Newton K, et al.: Lynch syndrome caused by MLH1 mutations is associated with an increased risk of breast cancer: a cohort study. J Med Genet 52 (8): 553-6, 2015. [PUBMED Abstract]
  343. Win AK, Lindor NM, Jenkins MA: Risk of breast cancer in Lynch syndrome: a systematic review. Breast Cancer Res 15 (2): R27, 2013. [PUBMED Abstract]
  344. Goldberg M, Bell K, Aronson M, et al.: Association between the Lynch syndrome gene MSH2 and breast cancer susceptibility in a Canadian familial cancer registry. J Med Genet 54 (11): 742-746, 2017. [PUBMED Abstract]
  345. Roberts ME, Jackson SA, Susswein LR, et al.: MSH6 and PMS2 germ-line pathogenic variants implicated in Lynch syndrome are associated with breast cancer. Genet Med 20 (10): 1167-1174, 2018. [PUBMED Abstract]
  346. Espenschied CR, LaDuca H, Li S, et al.: Multigene Panel Testing Provides a New Perspective on Lynch Syndrome. J Clin Oncol 35 (22): 2568-2575, 2017. [PUBMED Abstract]
  347. Lu HM, Li S, Black MH, et al.: Association of Breast and Ovarian Cancers With Predisposition Genes Identified by Large-Scale Sequencing. JAMA Oncol 5 (1): 51-57, 2019. [PUBMED Abstract]
  348. Latham A, Srinivasan P, Kemel Y, et al.: Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer. J Clin Oncol 37 (4): 286-295, 2019. [PUBMED Abstract]
  349. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 1.2021. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2020. Available online with free registration. Last accessed January 25, 2022.
  350. Harris CC, Hollstein M: Clinical implications of the p53 tumor-suppressor gene. N Engl J Med 329 (18): 1318-27, 1993. [PUBMED Abstract]
  351. Malkin D: The Li-Fraumeni syndrome. Cancer: Principles and Practice of Oncology Updates 7(7): 1-14, 1993.
  352. Gonzalez KD, Noltner KA, Buzin CH, et al.: Beyond Li Fraumeni Syndrome: clinical characteristics of families with p53 germline mutations. J Clin Oncol 27 (8): 1250-6, 2009. [PUBMED Abstract]
  353. Chompret A, Abel A, Stoppa-Lyonnet D, et al.: Sensitivity and predictive value of criteria for p53 germline mutation screening. J Med Genet 38 (1): 43-7, 2001. [PUBMED Abstract]
  354. Tinat J, Bougeard G, Baert-Desurmont S, et al.: 2009 version of the Chompret criteria for Li Fraumeni syndrome. J Clin Oncol 27 (26): e108-9; author reply e110, 2009. [PUBMED Abstract]
  355. Bottomley RH, Condit PT: Cancer families. Cancer Bull 20: 22-24, 1968.
  356. Bougeard G, Renaux-Petel M, Flaman JM, et al.: Revisiting Li-Fraumeni Syndrome From TP53 Mutation Carriers. J Clin Oncol 33 (21): 2345-52, 2015. [PUBMED Abstract]
  357. Sidransky D, Tokino T, Helzlsouer K, et al.: Inherited p53 gene mutations in breast cancer. Cancer Res 52 (10): 2984-6, 1992. [PUBMED Abstract]
  358. Wilson JR, Bateman AC, Hanson H, et al.: A novel HER2-positive breast cancer phenotype arising from germline TP53 mutations. J Med Genet 47 (11): 771-4, 2010. [PUBMED Abstract]
  359. Melhem-Bertrandt A, Bojadzieva J, Ready KJ, et al.: Early onset HER2-positive breast cancer is associated with germline TP53 mutations. Cancer 118 (4): 908-13, 2012. [PUBMED Abstract]
  360. Masciari S, Dillon DA, Rath M, et al.: Breast cancer phenotype in women with TP53 germline mutations: a Li-Fraumeni syndrome consortium effort. Breast Cancer Res Treat 133 (3): 1125-30, 2012. [PUBMED Abstract]
  361. Pearson AD, Craft AW, Ratcliffe JM, et al.: Two families with the Li-Fraumeni cancer family syndrome. J Med Genet 19 (5): 362-5, 1982. [PUBMED Abstract]
  362. Li FP, Fraumeni JF, Mulvihill JJ, et al.: A cancer family syndrome in twenty-four kindreds. Cancer Res 48 (18): 5358-62, 1988. [PUBMED Abstract]
  363. Bougeard G, Sesboüé R, Baert-Desurmont S, et al.: Molecular basis of the Li-Fraumeni syndrome: an update from the French LFS families. J Med Genet 45 (8): 535-8, 2008. [PUBMED Abstract]
  364. Mai PL, Best AF, Peters JA, et al.: Risks of first and subsequent cancers among TP53 mutation carriers in the National Cancer Institute Li-Fraumeni syndrome cohort. Cancer 122 (23): 3673-3681, 2016. [PUBMED Abstract]
  365. Kamihara J, Rana HQ, Garber JE: Germline TP53 mutations and the changing landscape of Li-Fraumeni syndrome. Hum Mutat 35 (6): 654-62, 2014. [PUBMED Abstract]
  366. Mai PL, Khincha PP, Loud JT, et al.: Prevalence of Cancer at Baseline Screening in the National Cancer Institute Li-Fraumeni Syndrome Cohort. JAMA Oncol 3 (12): 1640-1645, 2017. [PUBMED Abstract]
  367. Villani A, Tabori U, Schiffman J, et al.: Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: a prospective observational study. Lancet Oncol 12 (6): 559-67, 2011. [PUBMED Abstract]
  368. Masciari S, Van den Abbeele AD, Diller LR, et al.: F18-fluorodeoxyglucose-positron emission tomography/computed tomography screening in Li-Fraumeni syndrome. JAMA 299 (11): 1315-9, 2008. [PUBMED Abstract]
  369. Zhou XP, Waite KA, Pilarski R, et al.: Germline PTEN promoter mutations and deletions in Cowden/Bannayan-Riley-Ruvalcaba syndrome result in aberrant PTEN protein and dysregulation of the phosphoinositol-3-kinase/Akt pathway. Am J Hum Genet 73 (2): 404-11, 2003. [PUBMED Abstract]
  370. Mester J, Eng C: When overgrowth bumps into cancer: the PTEN-opathies. Am J Med Genet C Semin Med Genet 163C (2): 114-21, 2013. [PUBMED Abstract]
  371. Eng C: PTEN: one gene, many syndromes. Hum Mutat 22 (3): 183-98, 2003. [PUBMED Abstract]
  372. Marsh DJ, Kum JB, Lunetta KL, et al.: PTEN mutation spectrum and genotype-phenotype correlations in Bannayan-Riley-Ruvalcaba syndrome suggest a single entity with Cowden syndrome. Hum Mol Genet 8 (8): 1461-72, 1999. [PUBMED Abstract]
  373. Pilarski R, Eng C: Will the real Cowden syndrome please stand up (again)? Expanding mutational and clinical spectra of the PTEN hamartoma tumour syndrome. J Med Genet 41 (5): 323-6, 2004. [PUBMED Abstract]
  374. Eng C: PTEN Hamartoma Tumor Syndrome (PHTS). In: Pagon RA, Adam MP, Bird TD, et al., eds.: GeneReviews. Seattle, Wash: University of Washington, 1993-2018, pp. Available online. Last accessed April 1, 2022.
  375. Pilarski R, Burt R, Kohlman W, et al.: Cowden syndrome and the PTEN hamartoma tumor syndrome: systematic review and revised diagnostic criteria. J Natl Cancer Inst 105 (21): 1607-16, 2013. [PUBMED Abstract]
  376. Ngeow J, Liu C, Zhou K, et al.: Detecting Germline PTEN Mutations Among At-Risk Patients With Cancer: An Age- and Sex-Specific Cost-Effectiveness Analysis. J Clin Oncol 33 (23): 2537-44, 2015. [PUBMED Abstract]
  377. Tan MH, Mester JL, Ngeow J, et al.: Lifetime cancer risks in individuals with germline PTEN mutations. Clin Cancer Res 18 (2): 400-7, 2012. [PUBMED Abstract]
  378. Bubien V, Bonnet F, Brouste V, et al.: High cumulative risks of cancer in patients with PTEN hamartoma tumour syndrome. J Med Genet 50 (4): 255-63, 2013. [PUBMED Abstract]
  379. Myers MP, Tonks NK: PTEN: sometimes taking it off can be better than putting it on. Am J Hum Genet 61 (6): 1234-8, 1997. [PUBMED Abstract]
  380. Hobert JA, Eng C: PTEN hamartoma tumor syndrome: an overview. Genet Med 11 (10): 687-94, 2009. [PUBMED Abstract]
  381. Nieuwenhuis MH, Kets CM, Murphy-Ryan M, et al.: Cancer risk and genotype-phenotype correlations in PTEN hamartoma tumor syndrome. Fam Cancer 13 (1): 57-63, 2014. [PUBMED Abstract]
  382. Ngeow J, Stanuch K, Mester JL, et al.: Second malignant neoplasms in patients with Cowden syndrome with underlying germline PTEN mutations. J Clin Oncol 32 (17): 1818-24, 2014. [PUBMED Abstract]
  383. Olopade OI, Weber BL: Breast cancer genetics: toward molecular characterization of individuals at increased risk for breast cancer: part I. Cancer: Principles and Practice of Oncology Updates 12 (10): 1-12, 1998.
  384. Pilarski R, Stephens JA, Noss R, et al.: Predicting PTEN mutations: an evaluation of Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome clinical features. J Med Genet 48 (8): 505-12, 2011. [PUBMED Abstract]
  385. Nelen MR, Padberg GW, Peeters EA, et al.: Localization of the gene for Cowden disease to chromosome 10q22-23. Nat Genet 13 (1): 114-6, 1996. [PUBMED Abstract]
  386. Lachlan KL, Lucassen AM, Bunyan D, et al.: Cowden syndrome and Bannayan Riley Ruvalcaba syndrome represent one condition with variable expression and age-related penetrance: results of a clinical study of PTEN mutation carriers. J Med Genet 44 (9): 579-85, 2007. [PUBMED Abstract]
  387. Benusiglio PR, Malka D, Rouleau E, et al.: CDH1 germline mutations and the hereditary diffuse gastric and lobular breast cancer syndrome: a multicentre study. J Med Genet 50 (7): 486-9, 2013. [PUBMED Abstract]
  388. Beeghly-Fadiel A, Lu W, Gao YT, et al.: E-cadherin polymorphisms and breast cancer susceptibility: a report from the Shanghai Breast Cancer Study. Breast Cancer Res Treat 121 (2): 445-52, 2010. [PUBMED Abstract]
  389. McVeigh TP, Choi JK, Miller NM, et al.: Lobular breast cancer in a CDH1 splice site mutation carrier: case report and review of the literature. Clin Breast Cancer 14 (2): e47-51, 2014. [PUBMED Abstract]
  390. Petridis C, Shinomiya I, Kohut K, et al.: Germline CDH1 mutations in bilateral lobular carcinoma in situ. Br J Cancer 110 (4): 1053-7, 2014. [PUBMED Abstract]
  391. Tipirisetti NR, Govatati S, Govatati S, et al.: Association of E-cadherin single-nucleotide polymorphisms with the increased risk of breast cancer: a study in South Indian women. Genet Test Mol Biomarkers 17 (6): 494-500, 2013. [PUBMED Abstract]
  392. Fitzgerald RC, Hardwick R, Huntsman D, et al.: Hereditary diffuse gastric cancer: updated consensus guidelines for clinical management and directions for future research. J Med Genet 47 (7): 436-44, 2010. [PUBMED Abstract]
  393. Xie ZM, Li LS, Laquet C, et al.: Germline mutations of the E-cadherin gene in families with inherited invasive lobular breast carcinoma but no diffuse gastric cancer. Cancer 117 (14): 3112-7, 2011. [PUBMED Abstract]
  394. Peutz JL: Very remarkable case of familial polyposis of mucous membrane of intestinal tract and nasopharynx accompanied by peculiar pigmentations of skin and mucous membrane. Ned Tijdschr Geneeskd 10: 134-146, 1921.
  395. Jeghers H, McKusick VA, Katz KH: Generalized intestinal polyposis and melanin spots of the oral mucosa, lips and digits; a syndrome of diagnostic significance. N Engl J Med 241 (26): 1031-6, 1949. [PUBMED Abstract]
  396. Spigelman AD, Murday V, Phillips RK: Cancer and the Peutz-Jeghers syndrome. Gut 30 (11): 1588-90, 1989. [PUBMED Abstract]
  397. Aretz S, Stienen D, Uhlhaas S, et al.: High proportion of large genomic STK11 deletions in Peutz-Jeghers syndrome. Hum Mutat 26 (6): 513-9, 2005. [PUBMED Abstract]
  398. Hemminki A, Markie D, Tomlinson I, et al.: A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature 391 (6663): 184-7, 1998. [PUBMED Abstract]
  399. Jenne DE, Reimann H, Nezu J, et al.: Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet 18 (1): 38-43, 1998. [PUBMED Abstract]
  400. Boudeau J, Kieloch A, Alessi DR, et al.: Functional analysis of LKB1/STK11 mutants and two aberrant isoforms found in Peutz-Jeghers Syndrome patients. Hum Mutat 21 (2): 172, 2003. [PUBMED Abstract]
  401. Lim W, Hearle N, Shah B, et al.: Further observations on LKB1/STK11 status and cancer risk in Peutz-Jeghers syndrome. Br J Cancer 89 (2): 308-13, 2003. [PUBMED Abstract]
  402. Giardiello FM, Brensinger JD, Tersmette AC, et al.: Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 119 (6): 1447-53, 2000. [PUBMED Abstract]
  403. Hearle N, Schumacher V, Menko FH, et al.: Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res 12 (10): 3209-15, 2006. [PUBMED Abstract]
  404. Lim W, Olschwang S, Keller JJ, et al.: Relative frequency and morphology of cancers in STK11 mutation carriers. Gastroenterology 126 (7): 1788-94, 2004. [PUBMED Abstract]
  405. van Lier MG, Wagner A, Mathus-Vliegen EM, et al.: High cancer risk in Peutz-Jeghers syndrome: a systematic review and surveillance recommendations. Am J Gastroenterol 105 (6): 1258-64; author reply 1265, 2010. [PUBMED Abstract]
  406. Srivatsa PJ, Keeney GL, Podratz KC: Disseminated cervical adenoma malignum and bilateral ovarian sex cord tumors with annular tubules associated with Peutz-Jeghers syndrome. Gynecol Oncol 53 (2): 256-64, 1994. [PUBMED Abstract]
  407. Scully RE: Sex cord tumor with annular tubules a distinctive ovarian tumor of the Peutz-Jeghers syndrome. Cancer 25 (5): 1107-21, 1970. [PUBMED Abstract]
  408. Westerman AM, Entius MM, de Baar E, et al.: Peutz-Jeghers syndrome: 78-year follow-up of the original family. Lancet 353 (9160): 1211-5, 1999. [PUBMED Abstract]
  409. Mehenni H, Resta N, Park JG, et al.: Cancer risks in LKB1 germline mutation carriers. Gut 55 (7): 984-90, 2006. [PUBMED Abstract]
  410. Gruber SB, Entius MM, Petersen GM, et al.: Pathogenesis of adenocarcinoma in Peutz-Jeghers syndrome. Cancer Res 58 (23): 5267-70, 1998. [PUBMED Abstract]
  411. Wang ZJ, Ellis I, Zauber P, et al.: Allelic imbalance at the LKB1 (STK11) locus in tumours from patients with Peutz-Jeghers' syndrome provides evidence for a hamartoma-(adenoma)-carcinoma sequence. J Pathol 188 (1): 9-13, 1999. [PUBMED Abstract]
  412. Miyoshi H, Nakau M, Ishikawa TO, et al.: Gastrointestinal hamartomatous polyposis in Lkb1 heterozygous knockout mice. Cancer Res 62 (8): 2261-6, 2002. [PUBMED Abstract]
  413. Nakau M, Miyoshi H, Seldin MF, et al.: Hepatocellular carcinoma caused by loss of heterozygosity in Lkb1 gene knockout mice. Cancer Res 62 (16): 4549-53, 2002. [PUBMED Abstract]
  414. Takeda H, Miyoshi H, Kojima Y, et al.: Accelerated onsets of gastric hamartomas and hepatic adenomas/carcinomas in Lkb1+/-p53-/- compound mutant mice. Oncogene 25 (12): 1816-20, 2006. [PUBMED Abstract]
  415. Amos CI, Keitheri-Cheteri MB, Sabripour M, et al.: Genotype-phenotype correlations in Peutz-Jeghers syndrome. J Med Genet 41 (5): 327-33, 2004. [PUBMED Abstract]
  416. Reid S, Schindler D, Hanenberg H, et al.: Biallelic mutations in PALB2 cause Fanconi anemia subtype FA-N and predispose to childhood cancer. Nat Genet 39 (2): 162-4, 2007. [PUBMED Abstract]
  417. Rahman N, Seal S, Thompson D, et al.: PALB2, which encodes a BRCA2-interacting protein, is a breast cancer susceptibility gene. Nat Genet 39 (2): 165-7, 2007. [PUBMED Abstract]
  418. Erkko H, Dowty JG, Nikkilä J, et al.: Penetrance analysis of the PALB2 c.1592delT founder mutation. Clin Cancer Res 14 (14): 4667-71, 2008. [PUBMED Abstract]
  419. Heikkinen T, Kärkkäinen H, Aaltonen K, et al.: The breast cancer susceptibility mutation PALB2 1592delT is associated with an aggressive tumor phenotype. Clin Cancer Res 15 (9): 3214-22, 2009. [PUBMED Abstract]
  420. Ding YC, Steele L, Chu LH, et al.: Germline mutations in PALB2 in African-American breast cancer cases. Breast Cancer Res Treat 126 (1): 227-30, 2011. [PUBMED Abstract]
  421. Foulkes WD, Ghadirian P, Akbari MR, et al.: Identification of a novel truncating PALB2 mutation and analysis of its contribution to early-onset breast cancer in French-Canadian women. Breast Cancer Res 9 (6): R83, 2007. [PUBMED Abstract]
  422. Casadei S, Norquist BM, Walsh T, et al.: Contribution of inherited mutations in the BRCA2-interacting protein PALB2 to familial breast cancer. Cancer Res 71 (6): 2222-9, 2011. [PUBMED Abstract]
  423. Southey MC, Teo ZL, Dowty JG, et al.: A PALB2 mutation associated with high risk of breast cancer. Breast Cancer Res 12 (6): R109, 2010. [PUBMED Abstract]
  424. Hellebrand H, Sutter C, Honisch E, et al.: Germline mutations in the PALB2 gene are population specific and occur with low frequencies in familial breast cancer. Hum Mutat 32 (6): E2176-88, 2011. [PUBMED Abstract]
  425. Bogdanova N, Sokolenko AP, Iyevleva AG, et al.: PALB2 mutations in German and Russian patients with bilateral breast cancer. Breast Cancer Res Treat 126 (2): 545-50, 2011. [PUBMED Abstract]
  426. Wong MW, Nordfors C, Mossman D, et al.: BRIP1, PALB2, and RAD51C mutation analysis reveals their relative importance as genetic susceptibility factors for breast cancer. Breast Cancer Res Treat 127 (3): 853-9, 2011. [PUBMED Abstract]
  427. Zheng Y, Zhang J, Niu Q, et al.: Novel germline PALB2 truncating mutations in African American breast cancer patients. Cancer 118 (5): 1362-70, 2012. [PUBMED Abstract]
  428. Tischkowitz M, Capanu M, Sabbaghian N, et al.: Rare germline mutations in PALB2 and breast cancer risk: a population-based study. Hum Mutat 33 (4): 674-80, 2012. [PUBMED Abstract]
  429. Fernandes PH, Saam J, Peterson J, et al.: Comprehensive sequencing of PALB2 in patients with breast cancer suggests PALB2 mutations explain a subset of hereditary breast cancer. Cancer 120 (7): 963-7, 2014. [PUBMED Abstract]
  430. Janatova M, Kleibl Z, Stribrna J, et al.: The PALB2 gene is a strong candidate for clinical testing in BRCA1- and BRCA2-negative hereditary breast cancer. Cancer Epidemiol Biomarkers Prev 22 (12): 2323-32, 2013. [PUBMED Abstract]
  431. Teo ZL, Sawyer SD, James PA, et al.: The incidence of PALB2 c.3113G>A in women with a strong family history of breast and ovarian cancer attending familial cancer centres in Australia. Fam Cancer 12 (4): 587-95, 2013. [PUBMED Abstract]
  432. Snyder C, Metcalfe K, Sopik V, et al.: Prevalence of PALB2 mutations in the Creighton University Breast Cancer Family Registry. Breast Cancer Res Treat 150 (3): 637-41, 2015. [PUBMED Abstract]
  433. Nguyen-Dumont T, Hammet F, Mahmoodi M, et al.: Mutation screening of PALB2 in clinically ascertained families from the Breast Cancer Family Registry. Breast Cancer Res Treat 149 (2): 547-54, 2015. [PUBMED Abstract]
  434. Damiola F, Schultz I, Barjhoux L, et al.: Mutation analysis of PALB2 gene in French breast cancer families. Breast Cancer Res Treat 154 (3): 463-71, 2015. [PUBMED Abstract]
  435. Antoniou AC, Casadei S, Heikkinen T, et al.: Breast-cancer risk in families with mutations in PALB2. N Engl J Med 371 (6): 497-506, 2014. [PUBMED Abstract]
  436. Cybulski C, Kluźniak W, Huzarski T, et al.: Clinical outcomes in women with breast cancer and a PALB2 mutation: a prospective cohort analysis. Lancet Oncol 16 (6): 638-44, 2015. [PUBMED Abstract]
  437. Zhou J, Wang H, Fu F, et al.: Spectrum of PALB2 germline mutations and characteristics of PALB2-related breast cancer: Screening of 16,501 unselected patients with breast cancer and 5890 controls by next-generation sequencing. Cancer 126 (14): 3202-3208, 2020. [PUBMED Abstract]
  438. Yang X, Leslie G, Doroszuk A, et al.: Cancer Risks Associated With Germline PALB2 Pathogenic Variants: An International Study of 524 Families. J Clin Oncol 38 (7): 674-685, 2020. [PUBMED Abstract]
  439. Jones S, Hruban RH, Kamiyama M, et al.: Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science 324 (5924): 217, 2009. [PUBMED Abstract]
  440. Slater EP, Langer P, Niemczyk E, et al.: PALB2 mutations in European familial pancreatic cancer families. Clin Genet 78 (5): 490-4, 2010. [PUBMED Abstract]
  441. Hofstatter EW, Domchek SM, Miron A, et al.: PALB2 mutations in familial breast and pancreatic cancer. Fam Cancer 10 (2): 225-31, 2011. [PUBMED Abstract]
  442. Stadler ZK, Salo-Mullen E, Sabbaghian N, et al.: Germline PALB2 mutation analysis in breast-pancreas cancer families. J Med Genet 48 (8): 523-5, 2011. [PUBMED Abstract]
  443. Ghiorzo P, Pensotti V, Fornarini G, et al.: Contribution of germline mutations in the BRCA and PALB2 genes to pancreatic cancer in Italy. Fam Cancer 11 (1): 41-7, 2012. [PUBMED Abstract]
  444. Westerman AM, Entius MM, Boor PP, et al.: Novel mutations in the LKB1/STK11 gene in Dutch Peutz-Jeghers families. Hum Mutat 13 (6): 476-81, 1999. [PUBMED Abstract]
  445. Schreibman IR, Baker M, Amos C, et al.: The hamartomatous polyposis syndromes: a clinical and molecular review. Am J Gastroenterol 100 (2): 476-90, 2005. [PUBMED Abstract]
  446. Gonzalez KD, Buzin CH, Noltner KA, et al.: High frequency of de novo mutations in Li-Fraumeni syndrome. J Med Genet 46 (10): 689-93, 2009. [PUBMED Abstract]
  447. Bendig I, Mohr N, Kramer F, et al.: Identification of novel TP53 mutations in familial and sporadic cancer cases of German and Swiss origin. Cancer Genet Cytogenet 154 (1): 22-6, 2004. [PUBMED Abstract]
  448. De Leeneer K, Coene I, Crombez B, et al.: Prevalence of BRCA1/2 mutations in sporadic breast/ovarian cancer patients and identification of a novel de novo BRCA1 mutation in a patient diagnosed with late onset breast and ovarian cancer: implications for genetic testing. Breast Cancer Res Treat 132 (1): 87-95, 2012. [PUBMED Abstract]
  449. Diez O, Gutiérrez-Enríquez S, Mediano C, et al.: A novel de novo BRCA2 mutation of paternal origin identified in a Spanish woman with early onset bilateral breast cancer. Breast Cancer Res Treat 121 (1): 221-5, 2010. [PUBMED Abstract]
  450. Garcia-Casado Z, Romero I, Fernandez-Serra A, et al.: A de novo complete BRCA1 gene deletion identified in a Spanish woman with early bilateral breast cancer. BMC Med Genet 12: 134, 2011. [PUBMED Abstract]
  451. Hansen TV, Bisgaard ML, Jønson L, et al.: Novel de novo BRCA2 mutation in a patient with a family history of breast cancer. BMC Med Genet 9: 58, 2008. [PUBMED Abstract]
  452. Kwong A, Ng EK, Tang EY, et al.: A novel de novo BRCA1 mutation in a Chinese woman with early onset breast cancer. Fam Cancer 10 (2): 233-7, 2011. [PUBMED Abstract]
  453. Marshall M, Solomon S, Lawrence Wickerham D: Case report: de novo BRCA2 gene mutation in a 35-year-old woman with breast cancer. Clin Genet 76 (5): 427-30, 2009. [PUBMED Abstract]
  454. Robson M, Scheuer L, Nafa K, et al.: Unique de novo mutation of BRCA2 in a woman with early onset breast cancer. J Med Genet 39 (2): 126-8, 2002. [PUBMED Abstract]
  455. Tesoriero A, Andersen C, Southey M, et al.: De novo BRCA1 mutation in a patient with breast cancer and an inherited BRCA2 mutation. Am J Hum Genet 65 (2): 567-9, 1999. [PUBMED Abstract]
  456. van der Luijt RB, van Zon PH, Jansen RP, et al.: De novo recurrent germline mutation of the BRCA2 gene in a patient with early onset breast cancer. J Med Genet 38 (2): 102-5, 2001. [PUBMED Abstract]
  457. Morak M, Laner A, Scholz M, et al.: Report on de-novo mutation in the MSH2 gene as a rare event in hereditary nonpolyposis colorectal cancer. Eur J Gastroenterol Hepatol 20 (11): 1101-5, 2008. [PUBMED Abstract]
  458. Plasilova M, Zhang J, Okhowat R, et al.: A de novo MLH1 germ line mutation in a 31-year-old colorectal cancer patient. Genes Chromosomes Cancer 45 (12): 1106-10, 2006. [PUBMED Abstract]
  459. Win AK, Jenkins MA, Buchanan DD, et al.: Determining the frequency of de novo germline mutations in DNA mismatch repair genes. J Med Genet 48 (8): 530-4, 2011. [PUBMED Abstract]
  460. Anderson KG: How well does paternity confidence match actual paternity? Evidence from worldwide nonpaternity rates. Curr Anthropol 47 (3): 513-20, 2006. Also available online. Last accessed January 25, 2022.
  461. Sasse G, Müller H, Chakraborty R, et al.: Estimating the frequency of nonpaternity in Switzerland. Hum Hered 44 (6): 337-43, 1994 Nov-Dec. [PUBMED Abstract]
  462. Voracek M, Haubner T, Fisher ML: Recent decline in nonpaternity rates: a cross-temporal meta-analysis. Psychol Rep 103 (3): 799-811, 2008. [PUBMED Abstract]

Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer

Background

Pathogenic variants in BRCA1, BRCA2, PALB2, and the genes involved in other rare syndromes discussed in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary account for less than 25% of the familial risk of breast cancer.[1] Despite intensive genetic linkage studies, there do not appear to be other high-penetrance genes that account for a significant fraction of the remaining multiple-case familial clusters.[2] However, several moderate-penetrance genes associated with breast and/or gynecologic cancers have been identified. Genes such as CHEK2 and ATM are associated with a 20% or higher lifetime risk of breast cancer;[3,4] similarly, genes such as RAD51C, RAD51D, and BRIP1 are associated with a 5% to 10% risk of ovarian cancer.[5,6] Many of these genes are now included on multigene panels, although the clinical actionability of these findings remains uncertain and under investigation.

Breast and Gynecologic Cancer Susceptibility Genes Identified Through Candidate Gene Approaches

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

Fanconi anemia genes

Fanconi anemia (FA) is a rare, inherited condition characterized by bone marrow failure, increased risk of malignancy, and physical abnormalities. To date, 16 FA-related genes, including BRCA1 and BRCA2, have been identified (as outlined in Table 10). FA is mainly an autosomal recessive condition, except when caused by pathogenic variants in FANCB, which is X-linked recessive. FANCA accounts for 60% to 70% of pathogenic variants, FANCC accounts for approximately 14%, and the remaining genes each account for 3% or fewer.[7]

Table 10. Fanconi Anemia Genes and Breast Cancer Risk
aRefer to the BRCA1 and BRCA2 section of this summary for information about the cumulative risk of breast cancer in carriers of BRCA1 and BRCA2 pathogenic variants.
bRefer to the PALB2 section of this summary for information about the cumulative risk of breast cancer in carriers of PALB2 pathogenic variants.
cModerate risk is defined as a statistically significant, twofold or lower increased risk estimate.
High-Risk Genes
BRCA1 (FANCS)a
BRCA2 (FANCD1)a
PALB2 (FANCN)b
Moderate-Risk Genesc
BRIP1 (FANCJ/BACH1)
FANCD2
RAD51C (FANCO)
Genes With Uncertain or No Significantly Increased Risk
FANCA
FANCB
FANCC
FANCE
FANCF
FANCG (XRCC9)
FANCI (KIAA1794)
FANCL
SLX4 (FANCP)
ERCC4 (FANCQ/XPF)

Progressive bone marrow failure typically occurs in the first decade, with patients often presenting with thrombocytopenia or leucopenia. The incidence of bone marrow failure is 90% by age 40 to 50 years. The incidence is 10% to 30% for hematologic malignancies (primarily acute myeloid leukemia) and 25% to 30% for nonhematologic malignancies (solid tumors, particularly of the head and neck, skin, gastrointestinal [GI] tract, and genital tract). Physical abnormalities, including short stature, abnormal skin pigmentation, radial ray defects (including malformation of the thumbs), abnormalities of the urinary tract, eyes, ears, heart, GI system, and central nervous system, hypogonadism, and developmental delay are present in 60% to 75% of affected individuals.[7]

Variants in some of the FA genes, most notably BRCA1 and BRCA2, but also PALB2, RAD51C (in the RAD51 family of genes), and BRIP1, among others, may predispose to breast cancer in heterozygotes. Given the widespread availability of multigene (panel) tests, genetic testing of many of the FA genes is frequently performed despite uncertain cancer risks and the lack of available evidence-based medical management recommendations for many of these genes.

FA gene pathogenic variant carrier status can have implications for reproductive decision making because pathogenic variants in these genes can lead to serious childhood onset of disease if both parents are carriers of pathogenic variants in the same gene. Partner testing may be considered.

BRIP1

BRIP1 (also known as BACH1) encodes a helicase that interacts with the BRCA1 C-terminal (BRCT) domain. This gene also has a role in BRCA1-dependent DNA repair and cell cycle checkpoint function. Biallelic pathogenic variants in BRIP1 are a cause of FA,[8-10] much like such pathogenic variants in BRCA2. Inactivating variants of BRIP1 are associated with an increased risk of breast cancer. In one study, more than 3,000 individuals from BRCA1/BRCA2 pathogenic variant–negative families were examined for BRIP1 variants. Pathogenic variants were identified in 9 of 1,212 individuals with breast cancer but in only 2 of 2,081 controls (P = .003). The relative risk (RR) of breast cancer was estimated to be 2.0 (95% confidence interval [CI], 1.2–3.2; P = .012). Of note, in families with BRIP1 pathogenic variants and multiple cases of breast cancer, there was incomplete segregation of the pathogenic variant with breast cancer, consistent with a low-penetrance allele and similar to that seen with CHEK2.[11] In a case-control study of 3,236 women with ovarian cancer, BRIP1 pathogenic variants were more frequently associated with ovarian cancer risk (RR, 11.2; 95% CI, 3.2–34.1).[12]

CHEK2

CHEK2 is a gene involved in the DNA damage repair response pathway. Based on numerous studies, a polymorphism, 1100delC, appears to be a rare, moderate-penetrance cancer susceptibility allele.[13-18] One study identified the pathogenic variant in 1.2% of the European controls, 4.2% of the European BRCA1/BRCA2-negative familial breast cancer cases, and 1.4% of unselected female breast cancer cases.[13] In a group of 1,479 Dutch women younger than 50 years with invasive breast cancer, 3.7% were found to have the CHEK2 1100delC pathogenic variant.[19] In additional European and U.S. (where the pathogenic variant appears to be slightly less common) studies, including a large prospective study,[20] the frequency of CHEK2 pathogenic variants detected in familial breast or ovarian cancer cases has ranged from 0% [21] to 11%; overall, these studies have found an approximately 1.5-fold to 3-fold increased risk of female breast cancer.[20,22-25] A multicenter combined analysis and reanalysis of nearly 20,000 subjects from ten case-control studies, however, has verified a significant 2.3-fold excess of breast cancer among carriers of pathogenic variants.[26] A subsequent meta-analysis based on 29,154 cases and 37,064 controls from 25 case-control studies found a significant association between CHEK2 1100delC heterozygotes and breast cancer risk (odds ratio [OR], 2.75; 95% CI, 2.25–3.36). The ORs and CIs in unselected, familial, and early-onset breast cancer subgroups were 2.33 (1.79–3.05), 3.72 (2.61–5.31), and 2.78 (2.28–3.39), respectively. However, study limitations included pooling of populations without subgroup analysis, using a mix of population-based and hospital-based controls, and basing results on unadjusted estimates (as cases and controls were matched on only a few common factors); therefore, results should be interpreted in the context of these limitations.[27] In a series of male breast cancer patients, the CHEK2 1100delC variant was significantly more frequently identified than in controls, suggesting that this variant is also associated with an increased risk of male breast cancer.[28]

Two studies have suggested that the risk associated with a CHEK2 1100delC pathogenic variant was stronger in the families of probands ascertained because of bilateral breast cancer.[29,30] Furthermore, a meta-analysis of carriers of 1100delC pathogenic variants estimated the risk of breast cancer to be 42% by age 70 years in women with a family history of breast cancer.[31] Similarly, a Polish study reported that CHEK2 truncating pathogenic variants confer breast cancer risks based on a family history of breast cancer as follows: no family history: 20%; one second-degree relative: 28%; one first-degree relative: 34%; and both first- and second-degree relatives: 44%.[3] Moreover, a Dutch study suggested that female homozygotes for the CHEK2 1100delC variant have a greater-than-twofold increased breast cancer risk compared with heterozygotes.[32] Although there have been conflicting reports regarding cancers other than breast cancer associated with CHEK2 pathogenic variants, this may be dependent on variant type (i.e., missense vs. truncating) or population studied and is not currently of clinical utility.[18,23,33-38] The contribution of CHEK2 variants to breast cancer may depend on the population studied, with a potentially higher variant prevalence in Poland.[39] Carriers of CHEK2 variants in Poland may be more susceptible to estrogen receptor (ER)–positive breast cancer.[40]

Currently, the clinical applicability of CHEK variants remains uncertain because of low variant prevalence and lack of guidelines for clinical management.[41]

A large Dutch study of 86,975 individuals reported an increased risk of cancers other than breast and colon for carriers of the CHEK2 1100delC pathogenic variant,[42] although additional studies are needed to further refine these risks.

(Refer to the CHEK2 section in the PDQ summary on Genetics of Colorectal Cancer for more information.)

ATM

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by neurologic deterioration, telangiectasias, immunodeficiency states, and hypersensitivity to ionizing radiation. It is estimated that 1% of the general population may be heterozygote carriers of ATM variants.[43] More than 300 variants in the gene have been identified, most of which are truncating variants.[44] ATM proteins have been shown to play a role in cell cycle control.[45-47] In vitro, AT-deficient cells are sensitive to ionizing radiation and radiomimetic drugs, and lack cell cycle regulatory properties after exposure to radiation.[48] There is insufficient evidence to recommend against radiation therapy in carriers of a single ATM pathogenic variant (heterozygotes).

Initial studies searching for an excess of ATM pathogenic variants among breast cancer patients provided conflicting results, perhaps due to study design and variant testing strategies.[49-59] However, two large epidemiologic studies have demonstrated a statistically increased risk of breast cancer among female heterozygote carriers, with an estimated RR of approximately 2.0.[4,59] A meta-analysis modeled the risk of breast cancer to be 6.02% by age 50 years and 32.83% by age 80 years.[60] Given these risks, increased screening and other recommendations based on family history and age may be considered.

Some studies have shown an association between ATM and ovarian cancer,[61,62] although, at this time, there is no evidence to suggest an impact on risk management or disease characteristics.

CASP8 and TGFB1

The Breast Cancer Association Consortium (BCAC), an international group of investigators, investigated single nucleotide variants (SNVs) identified in previous studies as possibly associated with excess breast cancer risk in 15,000 to 20,000 cases and 15,000 to 20,000 controls. Two SNVs, CASP8 D302H and TGFB1 L10P, were associated with invasive breast cancer with RRs of 0.88 (95% CI, 0.84–0.92) and 1.08 (95% CI, 1.04–1.11), respectively.[63]

RAD51

RAD51 and the family of RAD51-related genes, also known as RAD51 paralogs, are thought to encode proteins that are involved in DNA damage repair through homologous recombination and interaction with numerous other DNA repair proteins, including BRCA1 and BRCA2. RAD51 protein plays a central role in single-strand annealing in the DNA damage response. RAD51 recruitment to break sites and recombinational DNA repair depend on the RAD51 paralogs, although their precise cellular functions are poorly characterized.[64] Variants in these genes are thought to result in loss of RAD51 focus formation in response to DNA damage.[65]

One of five RAD51-related genes, RAD51C has been reported to be linked to both FA-like disorders and familial breast and ovarian cancers. The literature, however, has produced contradictory findings. In a study of 480 German families characterized by breast and ovarian cancers who were negative for BRCA1 and BRCA2 pathogenic variants, six monoallelic variants in RAD51C were found (frequency of 1.3%).[66] Another study screened 286 BRCA1/BRCA2-negative patients with breast cancer and/or ovarian cancer and found one likely pathogenic variant in RAD51C-G153D.[67] RAD51C pathogenic variants have also been reported in Australian, British, Finnish, and Spanish non-BRCA1/BRCA2 ovarian cancer–only and breast/ovarian cancer families, and in unselected ovarian cancer cases, with frequencies ranging from 0% to 3% in these populations.[5,12,68-73] In a sample of 206 high-risk Jewish women (including 79 of Ashkenazi origin) previously tested for the common Jewish pathogenic variants, two previously described and possibly pathogenic missense variants were detected.[74] Four additional studies were unable to confirm an association between the RAD51C gene and hereditary breast cancer or ovarian cancer.[75-78]

In addition to carriers of RAD51C pathogenic variants, there are other RAD51 paralogs, including RAD51B, RAD51D, RAD51L1, XRCC2, and XRCC3, that may be associated with breast and/or ovarian cancer risk,[6,12,79-83] although the clinical significance of these findings is unknown. In a case-control study of 3,429 ovarian cancer patients, RAD51C and RAD51D pathogenic variants were more commonly found in ovarian cancer cases (0.82%) than in controls (0.11%, P < .001).[84]

In addition to germline variants, different polymorphisms of RAD51 have been hypothesized to have reduced capacity to repair DNA defects, resulting in increased susceptibility to familial breast cancer. The Consortium of Investigators of Modifiers of BRCA1/BRCA2 (CIMBA) pooled data from 8,512 carriers of BRCA1 and BRCA2 pathogenic variants and found evidence of an increased risk of breast cancer among women who were BRCA2 carriers and who were homozygous for CC at the RAD51 135G→C SNV (hazard ratio, 1.17; 95% CI, 0.91–1.51).[85]

Several meta-analyses have investigated the association between the RAD51 135G→C polymorphism and breast cancer risk. There is significant overlap in the studies reported in these meta-analyses, significant variability in the characteristics of the populations included, and significant methodologic limitations to their findings.[86-89] A meta-analysis of nine epidemiologic studies involving 13,241 cases and 13,203 controls of unknown BRCA1/BRCA2 status found that women carrying the CC genotype had an increased risk of breast cancer compared with women with the GG or GC genotype (OR, 1.35; 95% CI, 1.04–1.74). A meta-analysis of 14 case-control studies involving 12,183 cases and 10,183 controls confirmed an increased risk only for women who were known BRCA2 carriers (OR, 4.92; 95% CI, 1.10–21.83).[90] Another meta-analysis of 12 studies included only studies of known BRCA-negative cases and found no association between RAD51 135G→C and breast cancer.[91]

In summary, among this conflicting data is substantial evidence for a modest association between germline variants in RAD51C and breast cancer and ovarian cancer. There is also evidence of an association between polymorphisms in RAD51 135G→C among women with homozygous CC genotypes and breast cancer, particularly among BRCA2 carriers. These associations are plausible given the known role of RAD51 in the maintenance of genomic stability.

Abraxas

Pathogenic variants in the BRCA1-interacting gene Abraxas were found in three Finnish breast cancer families and no controls.[92] The significance of this finding outside of this population is not yet known.

RECQL

Through full exome sequencing among high-risk Polish and Quebec-based French Canadian families, the RECQL gene was discovered to harbor multiple rare truncating variants in both populations.[93] (Refer to the Clinical Sequencing section in the Cancer Genetics Overview PDQ summary for more information about whole-exome sequencing.) In the same populations, truncating variants in this gene were also identified in two subsequent validation phases among additional breast cancer patients from high-risk families, and among additional breast cancer cases in which the variant frequency was higher than that observed among controls. A case-control study from Belarus and Germany looked at the most common pathogenic variant, c.1667_1667+3delA GTA, and found it to be linked to ER-positive breast cancer. The OR in this study alone was 1.23 (95% CI, 0.44–3.47; P = .69), but in a meta-analysis with a Polish study, the OR was 2.51 (95% CI, 1.13–5.57, P = .02).[94] Although study results suggest that truncating germline RECQL pathogenic variants are associated with an increased risk of breast cancer, the exact magnitude of risk remains uncertain, and future studies are needed to determine clinical usefulness. Furthermore, the significance of this finding outside of these two populations is not yet known.

SMARCA4

SMARCA4 encodes BRG1 and is a catalytic subunit of the SWI/SNF chromatin remodeling complex, which plays a major role in rendering chromatin accessible to regulation of gene expression.

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, aggressive tumor that has an early age at onset (before age 40 y) and a poor prognosis.[95-97] Familial clustering is sometimes present. SCCOHT tumors may be unilateral or bilateral and have been characterized histologically by the presence of small hyperchromatic cells with brisk mitotic activity.[96] A multimodality approach including surgery, chemotherapy, and radiation therapy has been suggested for the treatment of SCCOHT.[96,97] Given the paraneoplastic phenomenon of hypercalcemia in 60% of cases, tracking calcium levels is useful in monitoring the course of disease. With a wide range of differential diagnoses including germ cell tumors, sex cord–stromal tumors, and undifferentiated carcinomas, SCCOHT remains classified by the World Health Organization as a "miscellaneous tumor" but more recently has been sequenced to be a malignant rhabdoid tumor.[98] Through exome sequencing, most cases of SCCOHT have been found to lack functional SMARCA4/BRG1; in fact, pathogenic variants in SMARCA4 may be the sole variants responsible for SCCOHT.

Despite only approximately 300 cases in the literature, three separate research groups showed SCCOHT to be associated with germline pathogenic variants and somatic mutations in the SMARCA4 gene. In one study of 12 young women with SCCOHT, sequencing of paired tumor and normal samples identified inactivating biallelic SMARCA4 pathogenic variants in each case.[99] Only four additional nonrecurrent somatic genes were identified in any of the other 278 genes sequenced. Immunohistochemistry demonstrated loss of SMARCA4 protein expression in seven of nine tested cases, consistent with a tumor-suppressor gene function. In a second study of another 12 patients, next-generation sequencing also identified SMARCA4 as the only recurrently variant gene, with the majority of variants predicted to result in a truncated protein.[100] A third study included three families in whom whole-exome sequencing with Sanger sequencing confirmation identified at least one germline pathogenic variant or somatic mutation in 24 of 26 cases.[101] Overall, 38 of 43 (88%) of SCCOHT tumors showed loss of SMARCA4 expression, in comparison to only 1 of 139 (0.7%) other ovarian tumor types.

Because of the rarity of this tumor, the penetrance of SMARCA4 is unknown. There is currently no consensus for management, yet SMARCA4 is on the larger multigene panels currently available for genetic testing, and risk-reducing surgery has been offered to pathogenic variant carriers.[102]

References
  1. Easton DF: How many more breast cancer predisposition genes are there? Breast Cancer Res 1 (1): 14-7, 1999. [PUBMED Abstract]
  2. Smith P, McGuffog L, Easton DF, et al.: A genome wide linkage search for breast cancer susceptibility genes. Genes Chromosomes Cancer 45 (7): 646-55, 2006. [PUBMED Abstract]
  3. Cybulski C, Wokołorczyk D, Jakubowska A, et al.: Risk of breast cancer in women with a CHEK2 mutation with and without a family history of breast cancer. J Clin Oncol 29 (28): 3747-52, 2011. [PUBMED Abstract]
  4. Thompson D, Duedal S, Kirner J, et al.: Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst 97 (11): 813-22, 2005. [PUBMED Abstract]
  5. Pelttari LM, Heikkinen T, Thompson D, et al.: RAD51C is a susceptibility gene for ovarian cancer. Hum Mol Genet 20 (16): 3278-88, 2011. [PUBMED Abstract]
  6. Loveday C, Turnbull C, Ramsay E, et al.: Germline mutations in RAD51D confer susceptibility to ovarian cancer. Nat Genet 43 (9): 879-82, 2011. [PUBMED Abstract]
  7. Mehta PA, Tolar J: Fanconi Anemia. In: Adam MP, Ardinger HH, Pagon RA, et al.: GeneReviews. Seattle, Wash: University of Washington, 1993-2018, pp. Available online. Last accessed February 4, 2022.
  8. Levitus M, Waisfisz Q, Godthelp BC, et al.: The DNA helicase BRIP1 is defective in Fanconi anemia complementation group J. Nat Genet 37 (9): 934-5, 2005. [PUBMED Abstract]
  9. Levran O, Attwooll C, Henry RT, et al.: The BRCA1-interacting helicase BRIP1 is deficient in Fanconi anemia. Nat Genet 37 (9): 931-3, 2005. [PUBMED Abstract]
  10. Litman R, Peng M, Jin Z, et al.: BACH1 is critical for homologous recombination and appears to be the Fanconi anemia gene product FANCJ. Cancer Cell 8 (3): 255-65, 2005. [PUBMED Abstract]
  11. Seal S, Thompson D, Renwick A, et al.: Truncating mutations in the Fanconi anemia J gene BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat Genet 38 (11): 1239-41, 2006. [PUBMED Abstract]
  12. Ramus SJ, Song H, Dicks E, et al.: Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer. J Natl Cancer Inst 107 (11): , 2015. [PUBMED Abstract]
  13. Meijers-Heijboer H, van den Ouweland A, Klijn J, et al.: Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nat Genet 31 (1): 55-9, 2002. [PUBMED Abstract]
  14. Kuschel B, Auranen A, Gregory CS, et al.: Common polymorphisms in checkpoint kinase 2 are not associated with breast cancer risk. Cancer Epidemiol Biomarkers Prev 12 (8): 809-12, 2003. [PUBMED Abstract]
  15. Sodha N, Bullock S, Taylor R, et al.: CHEK2 variants in susceptibility to breast cancer and evidence of retention of the wild type allele in tumours. Br J Cancer 87 (12): 1445-8, 2002. [PUBMED Abstract]
  16. Ingvarsson S, Sigbjornsdottir BI, Huiping C, et al.: Mutation analysis of the CHK2 gene in breast carcinoma and other cancers. Breast Cancer Res 4 (3): R4, 2002. [PUBMED Abstract]
  17. Vahteristo P, Bartkova J, Eerola H, et al.: A CHEK2 genetic variant contributing to a substantial fraction of familial breast cancer. Am J Hum Genet 71 (2): 432-8, 2002. [PUBMED Abstract]
  18. Meijers-Heijboer H, Wijnen J, Vasen H, et al.: The CHEK2 1100delC mutation identifies families with a hereditary breast and colorectal cancer phenotype. Am J Hum Genet 72 (5): 1308-14, 2003. [PUBMED Abstract]
  19. Schmidt MK, Tollenaar RA, de Kemp SR, et al.: Breast cancer survival and tumor characteristics in premenopausal women carrying the CHEK2*1100delC germline mutation. J Clin Oncol 25 (1): 64-9, 2007. [PUBMED Abstract]
  20. Weischer M, Bojesen SE, Tybjaerg-Hansen A, et al.: Increased risk of breast cancer associated with CHEK2*1100delC. J Clin Oncol 25 (1): 57-63, 2007. [PUBMED Abstract]
  21. Iniesta MD, Gorin MA, Chien LC, et al.: Absence of CHEK2*1100delC mutation in families with hereditary breast cancer in North America. Cancer Genet Cytogenet 202 (2): 136-40, 2010. [PUBMED Abstract]
  22. Offit K, Pierce H, Kirchhoff T, et al.: Frequency of CHEK2*1100delC in New York breast cancer cases and controls. BMC Med Genet 4 (1): 1, 2003. [PUBMED Abstract]
  23. Oldenburg RA, Kroeze-Jansema K, Kraan J, et al.: The CHEK2*1100delC variant acts as a breast cancer risk modifier in non-BRCA1/BRCA2 multiple-case families. Cancer Res 63 (23): 8153-7, 2003. [PUBMED Abstract]
  24. Neuhausen S, Dunning A, Steele L, et al.: Role of CHEK2*1100delC in unselected series of non-BRCA1/2 male breast cancers. Int J Cancer 108 (3): 477-8, 2004. [PUBMED Abstract]
  25. Ohayon T, Gal I, Baruch RG, et al.: CHEK2*1100delC and male breast cancer risk in Israel. Int J Cancer 108 (3): 479-80, 2004. [PUBMED Abstract]
  26. CHEK2 Breast Cancer Case-Control Consortium: CHEK2*1100delC and susceptibility to breast cancer: a collaborative analysis involving 10,860 breast cancer cases and 9,065 controls from 10 studies. Am J Hum Genet 74 (6): 1175-82, 2004. [PUBMED Abstract]
  27. Yang Y, Zhang F, Wang Y, et al.: CHEK2 1100delC variant and breast cancer risk in Caucasians: a meta-analysis based on 25 studies with 29,154 cases and 37,064 controls. Asian Pac J Cancer Prev 13 (7): 3501-5, 2012. [PUBMED Abstract]
  28. Hallamies S, Pelttari LM, Poikonen-Saksela P, et al.: CHEK2 c.1100delC mutation is associated with an increased risk for male breast cancer in Finnish patient population. BMC Cancer 17 (1): 620, 2017. [PUBMED Abstract]
  29. Johnson N, Fletcher O, Naceur-Lombardelli C, et al.: Interaction between CHEK2*1100delC and other low-penetrance breast-cancer susceptibility genes: a familial study. Lancet 366 (9496): 1554-7, 2005 Oct 29-Nov 4. [PUBMED Abstract]
  30. Fletcher O, Johnson N, Dos Santos Silva I, et al.: Family history, genetic testing, and clinical risk prediction: pooled analysis of CHEK2 1100delC in 1,828 bilateral breast cancers and 7,030 controls. Cancer Epidemiol Biomarkers Prev 18 (1): 230-4, 2009. [PUBMED Abstract]
  31. Weischer M, Bojesen SE, Ellervik C, et al.: CHEK2*1100delC genotyping for clinical assessment of breast cancer risk: meta-analyses of 26,000 patient cases and 27,000 controls. J Clin Oncol 26 (4): 542-8, 2008. [PUBMED Abstract]
  32. Adank MA, Jonker MA, Kluijt I, et al.: CHEK2*1100delC homozygosity is associated with a high breast cancer risk in women. J Med Genet 48 (12): 860-3, 2011. [PUBMED Abstract]
  33. Gronwald J, Cybulski C, Piesiak W, et al.: Cancer risks in first-degree relatives of CHEK2 mutation carriers: effects of mutation type and cancer site in proband. Br J Cancer 100 (9): 1508-12, 2009. [PUBMED Abstract]
  34. Wasielewski M, den Bakker MA, van den Ouweland A, et al.: CHEK2 1100delC and male breast cancer in the Netherlands. Breast Cancer Res Treat 116 (2): 397-400, 2009. [PUBMED Abstract]
  35. Osorio A, Rodríguez-López R, Díez O, et al.: The breast cancer low-penetrance allele 1100delC in the CHEK2 gene is not present in Spanish familial breast cancer population. Int J Cancer 108 (1): 54-6, 2004. [PUBMED Abstract]
  36. Syrjäkoski K, Kuukasjärvi T, Auvinen A, et al.: CHEK2 1100delC is not a risk factor for male breast cancer population. Int J Cancer 108 (3): 475-6, 2004. [PUBMED Abstract]
  37. Tsou HC, Teng DH, Ping XL, et al.: The role of MMAC1 mutations in early-onset breast cancer: causative in association with Cowden syndrome and excluded in BRCA1-negative cases. Am J Hum Genet 61 (5): 1036-43, 1997. [PUBMED Abstract]
  38. Olopade OI, Weber BL: Breast cancer genetics: toward molecular characterization of individuals at increased risk for breast cancer: part I. Cancer: Principles and Practice of Oncology Updates 12 (10): 1-12, 1998.
  39. Cybulski C, Górski B, Huzarski T, et al.: CHEK2-positive breast cancers in young Polish women. Clin Cancer Res 12 (16): 4832-5, 2006. [PUBMED Abstract]
  40. Cybulski C, Huzarski T, Byrski T, et al.: Estrogen receptor status in CHEK2-positive breast cancers: implications for chemoprevention. Clin Genet 75 (1): 72-8, 2009. [PUBMED Abstract]
  41. Offit K, Garber JE: Time to check CHEK2 in families with breast cancer? J Clin Oncol 26 (4): 519-20, 2008. [PUBMED Abstract]
  42. Näslund-Koch C, Nordestgaard BG, Bojesen SE: Increased Risk for Other Cancers in Addition to Breast Cancer for CHEK2*1100delC Heterozygotes Estimated From the Copenhagen General Population Study. J Clin Oncol 34 (11): 1208-16, 2016. [PUBMED Abstract]
  43. Savitsky K, Bar-Shira A, Gilad S, et al.: A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268 (5218): 1749-53, 1995. [PUBMED Abstract]
  44. Telatar M, Teraoka S, Wang Z, et al.: Ataxia-telangiectasia: identification and detection of founder-effect mutations in the ATM gene in ethnic populations. Am J Hum Genet 62 (1): 86-97, 1998. [PUBMED Abstract]
  45. Uhrhammer N, Bay JO, Bignon YJ: Seventh International Workshop on Ataxia-Telangiectasia. Cancer Res 58 (15): 3480-5, 1998. [PUBMED Abstract]
  46. Ahmed M, Rahman N: ATM and breast cancer susceptibility. Oncogene 25 (43): 5906-11, 2006. [PUBMED Abstract]
  47. Khanna KK, Chenevix-Trench G: ATM and genome maintenance: defining its role in breast cancer susceptibility. J Mammary Gland Biol Neoplasia 9 (3): 247-62, 2004. [PUBMED Abstract]
  48. Gilad S, Chessa L, Khosravi R, et al.: Genotype-phenotype relationships in ataxia-telangiectasia and variants. Am J Hum Genet 62 (3): 551-61, 1998. [PUBMED Abstract]
  49. FitzGerald MG, Bean JM, Hegde SR, et al.: Heterozygous ATM mutations do not contribute to early onset of breast cancer. Nat Genet 15 (3): 307-10, 1997. [PUBMED Abstract]
  50. Chen J, Birkholtz GG, Lindblom P, et al.: The role of ataxia-telangiectasia heterozygotes in familial breast cancer. Cancer Res 58 (7): 1376-9, 1998. [PUBMED Abstract]
  51. Bay JO, Grancho M, Pernin D, et al.: No evidence for constitutional ATM mutation in breast/gastric cancer families. Int J Oncol 12 (6): 1385-90, 1998. [PUBMED Abstract]
  52. Laake K, Vu P, Andersen TI, et al.: Screening breast cancer patients for Norwegian ATM mutations. Br J Cancer 83 (12): 1650-3, 2000. [PUBMED Abstract]
  53. Dörk T, Bendix R, Bremer M, et al.: Spectrum of ATM gene mutations in a hospital-based series of unselected breast cancer patients. Cancer Res 61 (20): 7608-15, 2001. [PUBMED Abstract]
  54. Teraoka SN, Malone KE, Doody DR, et al.: Increased frequency of ATM mutations in breast carcinoma patients with early onset disease and positive family history. Cancer 92 (3): 479-87, 2001. [PUBMED Abstract]
  55. Chenevix-Trench G, Spurdle AB, Gatei M, et al.: Dominant negative ATM mutations in breast cancer families. J Natl Cancer Inst 94 (3): 205-15, 2002. [PUBMED Abstract]
  56. Thorstenson YR, Roxas A, Kroiss R, et al.: Contributions of ATM mutations to familial breast and ovarian cancer. Cancer Res 63 (12): 3325-33, 2003. [PUBMED Abstract]
  57. Cavaciuti E, Laugé A, Janin N, et al.: Cancer risk according to type and location of ATM mutation in ataxia-telangiectasia families. Genes Chromosomes Cancer 42 (1): 1-9, 2005. [PUBMED Abstract]
  58. Olsen JH, Hahnemann JM, Børresen-Dale AL, et al.: Breast and other cancers in 1445 blood relatives of 75 Nordic patients with ataxia telangiectasia. Br J Cancer 93 (2): 260-5, 2005. [PUBMED Abstract]
  59. Renwick A, Thompson D, Seal S, et al.: ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet 38 (8): 873-5, 2006. [PUBMED Abstract]
  60. Marabelli M, Cheng SC, Parmigiani G: Penetrance of ATM Gene Mutations in Breast Cancer: A Meta-Analysis of Different Measures of Risk. Genet Epidemiol 40 (5): 425-31, 2016. [PUBMED Abstract]
  61. Lu HM, Li S, Black MH, et al.: Association of Breast and Ovarian Cancers With Predisposition Genes Identified by Large-Scale Sequencing. JAMA Oncol 5 (1): 51-57, 2019. [PUBMED Abstract]
  62. Pennington KP, Walsh T, Harrell MI, et al.: Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin Cancer Res 20 (3): 764-75, 2014. [PUBMED Abstract]
  63. Cox Angela, Dunning Alison, Garcia-Closas Montserrat, et al.: Nature genetics. Nat Genet 39 (5): 352-8, 2007.
  64. Suwaki N, Klare K, Tarsounas M: RAD51 paralogs: roles in DNA damage signalling, recombinational repair and tumorigenesis. Semin Cell Dev Biol 22 (8): 898-905, 2011. [PUBMED Abstract]
  65. Vaz F, Hanenberg H, Schuster B, et al.: Mutation of the RAD51C gene in a Fanconi anemia-like disorder. Nat Genet 42 (5): 406-9, 2010. [PUBMED Abstract]
  66. Meindl A, Hellebrand H, Wiek C, et al.: Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene. Nat Genet 42 (5): 410-4, 2010. [PUBMED Abstract]
  67. Clague J, Wilhoite G, Adamson A, et al.: RAD51C germline mutations in breast and ovarian cancer cases from high-risk families. PLoS One 6 (9): e25632, 2011. [PUBMED Abstract]
  68. Thompson ER, Boyle SE, Johnson J, et al.: Analysis of RAD51C germline mutations in high-risk breast and ovarian cancer families and ovarian cancer patients. Hum Mutat 33 (1): 95-9, 2012. [PUBMED Abstract]
  69. Vuorela M, Pylkäs K, Hartikainen JM, et al.: Further evidence for the contribution of the RAD51C gene in hereditary breast and ovarian cancer susceptibility. Breast Cancer Res Treat 130 (3): 1003-10, 2011. [PUBMED Abstract]
  70. Romero A, Pérez-Segura P, Tosar A, et al.: A HRM-based screening method detects RAD51C germ-line deleterious mutations in Spanish breast and ovarian cancer families. Breast Cancer Res Treat 129 (3): 939-46, 2011. [PUBMED Abstract]
  71. Osorio A, Endt D, Fernández F, et al.: Predominance of pathogenic missense variants in the RAD51C gene occurring in breast and ovarian cancer families. Hum Mol Genet 21 (13): 2889-98, 2012. [PUBMED Abstract]
  72. Blanco A, Gutiérrez-Enríquez S, Santamariña M, et al.: RAD51C germline mutations found in Spanish site-specific breast cancer and breast-ovarian cancer families. Breast Cancer Res Treat 147 (1): 133-43, 2014. [PUBMED Abstract]
  73. Norquist BM, Harrell MI, Brady MF, et al.: Inherited Mutations in Women With Ovarian Carcinoma. JAMA Oncol 2 (4): 482-90, 2016. [PUBMED Abstract]
  74. Kushnir A, Laitman Y, Shimon SP, et al.: Germline mutations in RAD51C in Jewish high cancer risk families. Breast Cancer Res Treat 136 (3): 869-74, 2012. [PUBMED Abstract]
  75. Wong MW, Nordfors C, Mossman D, et al.: BRIP1, PALB2, and RAD51C mutation analysis reveals their relative importance as genetic susceptibility factors for breast cancer. Breast Cancer Res Treat 127 (3): 853-9, 2011. [PUBMED Abstract]
  76. Zheng Y, Zhang J, Hope K, et al.: Screening RAD51C nucleotide alterations in patients with a family history of breast and ovarian cancer. Breast Cancer Res Treat 124 (3): 857-61, 2010. [PUBMED Abstract]
  77. Akbari MR, Tonin P, Foulkes WD, et al.: RAD51C germline mutations in breast and ovarian cancer patients. Breast Cancer Res 12 (4): 404, 2010. [PUBMED Abstract]
  78. De Leeneer K, Van Bockstal M, De Brouwer S, et al.: Evaluation of RAD51C as cancer susceptibility gene in a large breast-ovarian cancer patient population referred for genetic testing. Breast Cancer Res Treat 133 (1): 393-8, 2012. [PUBMED Abstract]
  79. Thomas G, Jacobs KB, Kraft P, et al.: A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11.2 and 14q24.1 (RAD51L1). Nat Genet 41 (5): 579-84, 2009. [PUBMED Abstract]
  80. Figueroa JD, Garcia-Closas M, Humphreys M, et al.: Associations of common variants at 1p11.2 and 14q24.1 (RAD51L1) with breast cancer risk and heterogeneity by tumor subtype: findings from the Breast Cancer Association Consortium. Hum Mol Genet 20 (23): 4693-706, 2011. [PUBMED Abstract]
  81. Osher DJ, De Leeneer K, Michils G, et al.: Mutation analysis of RAD51D in non-BRCA1/2 ovarian and breast cancer families. Br J Cancer 106 (8): 1460-3, 2012. [PUBMED Abstract]
  82. Pelttari LM, Kiiski J, Nurminen R, et al.: A Finnish founder mutation in RAD51D: analysis in breast, ovarian, prostate, and colorectal cancer. J Med Genet 49 (7): 429-32, 2012. [PUBMED Abstract]
  83. Golmard L, Castéra L, Krieger S, et al.: Contribution of germline deleterious variants in the RAD51 paralogs to breast and ovarian cancers. Eur J Hum Genet 25 (12): 1345-1353, 2017. [PUBMED Abstract]
  84. Song H, Dicks E, Ramus SJ, et al.: Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population. J Clin Oncol 33 (26): 2901-7, 2015. [PUBMED Abstract]
  85. Antoniou AC, Sinilnikova OM, Simard J, et al.: RAD51 135G-->C modifies breast cancer risk among BRCA2 mutation carriers: results from a combined analysis of 19 studies. Am J Hum Genet 81 (6): 1186-200, 2007. [PUBMED Abstract]
  86. He XF, Su J, Zhang Y, et al.: Need for clarification of data in the recent meta-analysis about RAD51 135G>C polymorphism and breast cancer risk. Breast Cancer Res Treat 129 (2): 649-51; author reply 652-3, 2011. [PUBMED Abstract]
  87. Lu W, Wang X, Lin H, et al.: Mutation screening of RAD51C in high-risk breast and ovarian cancer families. Fam Cancer 11 (3): 381-5, 2012. [PUBMED Abstract]
  88. Wang WW, Spurdle AB, Kolachana P, et al.: A single nucleotide polymorphism in the 5' untranslated region of RAD51 and risk of cancer among BRCA1/2 mutation carriers. Cancer Epidemiol Biomarkers Prev 10 (9): 955-60, 2001. [PUBMED Abstract]
  89. Wang Z, Dong H, Fu Y, et al.: RAD51 135G>C polymorphism contributes to breast cancer susceptibility: a meta-analysis involving 26,444 subjects. Breast Cancer Res Treat 124 (3): 765-9, 2010. [PUBMED Abstract]
  90. Zhou GW, Hu J, Peng XD, et al.: RAD51 135G>C polymorphism and breast cancer risk: a meta-analysis. Breast Cancer Res Treat 125 (2): 529-35, 2011. [PUBMED Abstract]
  91. Yu KD, Yang C, Fan L, et al.: RAD51 135G>C does not modify breast cancer risk in non-BRCA1/2 mutation carriers: evidence from a meta-analysis of 12 studies. Breast Cancer Res Treat 126 (2): 365-71, 2011. [PUBMED Abstract]
  92. Solyom S, Aressy B, Pylkäs K, et al.: Breast cancer-associated Abraxas mutation disrupts nuclear localization and DNA damage response functions. Sci Transl Med 4 (122): 122ra23, 2012. [PUBMED Abstract]
  93. Cybulski C, Carrot-Zhang J, Kluźniak W, et al.: Germline RECQL mutations are associated with breast cancer susceptibility. Nat Genet 47 (6): 643-6, 2015. [PUBMED Abstract]
  94. Bogdanova N, Pfeifer K, Schürmann P, et al.: Analysis of a RECQL splicing mutation, c.1667_1667+3delAGTA, in breast cancer patients and controls from Central Europe. Fam Cancer 16 (2): 181-186, 2017. [PUBMED Abstract]
  95. Dickersin GR, Kline IW, Scully RE: Small cell carcinoma of the ovary with hypercalcemia: a report of eleven cases. Cancer 49 (1): 188-97, 1982. [PUBMED Abstract]
  96. Harrison ML, Hoskins P, du Bois A, et al.: Small cell of the ovary, hypercalcemic type -- analysis of combined experience and recommendation for management. A GCIG study. Gynecol Oncol 100 (2): 233-8, 2006. [PUBMED Abstract]
  97. Callegaro-Filho D, Gershenson DM, Nick AM, et al.: Small cell carcinoma of the ovary-hypercalcemic type (SCCOHT): A review of 47 cases. Gynecol Oncol 140 (1): 53-7, 2016. [PUBMED Abstract]
  98. Foulkes WD, Clarke BA, Hasselblatt M, et al.: No small surprise - small cell carcinoma of the ovary, hypercalcaemic type, is a malignant rhabdoid tumour. J Pathol 233 (3): 209-14, 2014. [PUBMED Abstract]
  99. Jelinic P, Mueller JJ, Olvera N, et al.: Recurrent SMARCA4 mutations in small cell carcinoma of the ovary. Nat Genet 46 (5): 424-6, 2014. [PUBMED Abstract]
  100. Ramos P, Karnezis AN, Craig DW, et al.: Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4. Nat Genet 46 (5): 427-9, 2014. [PUBMED Abstract]
  101. Witkowski L, Carrot-Zhang J, Albrecht S, et al.: Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nat Genet 46 (5): 438-43, 2014. [PUBMED Abstract]
  102. Berchuck A, Witkowski L, Hasselblatt M, et al.: Prophylactic oophorectomy for hereditary small cell carcinoma of the ovary, hypercalcemic type. Gynecol Oncol Rep 12: 20-2, 2015. [PUBMED Abstract]

Single Nucleotide Variant–Associated Cancer Risks

Polymorphisms underlying polygenic susceptibility to breast and gynecologic cancers are considered low penetrance, a term often applied to sequence variants associated with a minimal to moderate risk. This is in contrast to high-penetrance variants or alleles that are typically associated with more severe phenotypes, for example BRCA1/BRCA2 pathogenic variants leading to an autosomal dominant inheritance pattern in a family, and moderate-penetrance variants such as BRIP1, CHEK2, and RAD51C. (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes and the Moderate-Penetrance Genes Associated With Breast and/or Gynecologic Cancer sections of this summary for more information.) Because these types of sequence variants (also called low-penetrance genes, alleles, variants, and polymorphisms) are relatively common in the general population, their overall contribution to cancer risk is estimated to be much greater than the attributable risk in the population from pathogenic variants in BRCA1 and BRCA2. For example, it is estimated by segregation analysis that half of all breast cancer occurs in 12% of the population that is deemed most susceptible.[1] There are no known low-penetrance variants in BRCA1/BRCA2. The N372H variation in BRCA2, initially thought to be a low-penetrance allele, was not verified in a large combined analysis.[2]

Two strategies have attempted to identify low-penetrance polymorphisms leading to breast cancer susceptibility: candidate gene and genome-wide searches. Both involve the epidemiologic case-control study design. The candidate gene approach involves selecting genes based on their known or presumed biological function, relevance to carcinogenesis or organ physiology, and then searching for or testing known genetic variants for an association with cancer risk. This strategy relies on imperfect and incomplete biological knowledge, and, despite some confirmed associations (described below), has been relatively disappointing.[2,3] The candidate gene approach has largely been replaced by genome-wide association studies (GWAS) in which a very large number of single nucleotide variants (SNVs) (approximately 1 million to 5 million) are chosen within the genome and tested, mostly without regard to their possible biological function, but instead to more uniformly capture all genetic variation throughout the genome.

Genome-Wide Searches

In contrast to assessing candidate genes and/or alleles, GWAS involve comparing a very large set of genetic variants spread throughout the genome. The current paradigm uses sets of as many as 5 million SNVs that are chosen to capture a large portion of common variation within the genome based on the HapMap and the 1000 Genomes Project.[4,5] By comparing allele frequencies between a large number of cases and controls, typically 1,000 or more of each, and validating promising signals in replication sets of subjects, very robust statistical signals of association have been obtained.[6-8] The strong correlation between many SNVs that are physically close to each other on the chromosome (linkage disequilibrium) allows one to “scan” the genome for susceptibility alleles even if the biologically relevant variant is not within the tested set of SNVs. Although this between-SNV correlation allows one to interrogate the majority of the genome without having to assay every SNV, when a validated association is obtained, it is not usually obvious which of the many correlated variants is causal.

Genome-wide searches are showing great promise in identifying common, low-penetrance susceptibility alleles for many complex diseases,[9] including breast cancer.[10-13] The first study involved an initial scan in familial breast cancer cases followed by replication in two large sample sets of sporadic breast cancer, the final being a collection of over 20,000 cases and 20,000 controls from the Breast Cancer Association Consortium.[10] Five distinct genomic regions were identified that were within or near the FGFR2, TNRC9, MAP3K1, and LSP1 genes or at the chromosome 8q region. The 8q region and others may harbor multiple independent loci associated with risk. Subsequent genome-wide studies have replicated these loci and identified additional ones.[11,12,14,14-19] Numerous SNVs identified through large studies of sporadic breast cancer appear to be associated more strongly with estrogen receptor (ER)–positive disease;[20] however, some are associated primarily or exclusively with other subtypes, including triple-negative disease.[21,22] An online catalog is available of SNV-trait associations from published GWAS for use in investigating genomic characteristics of trait/disease-associated SNVs.

Although the statistical evidence for an association between genetic variation at these loci and breast and ovarian cancer risk is overwhelming, the biologically relevant variants and the mechanism by which they lead to increased risk are unknown and will require further genetic and functional characterization. Additionally, these loci are associated with very modest risk (typically, an odds ratio [OR] <1.5), with more risk variants likely to be identified. No interaction between the SNVs and epidemiologic risk factors for breast cancer have been identified.[23,24] Furthermore, theoretical models have suggested that common moderate-risk SNVs have limited potential to improve models for individualized risk assessment.[25-27] These models used receiver operating characteristic (ROC) curve analysis to calculate the area under the curve (AUC) as a measure of discriminatory accuracy. A subsequent study used ROC curve analysis to examine the utility of SNVs in a clinical dataset of more than 5,500 breast cancer cases and nearly 6,000 controls, using a model with traditional risk factors compared with a model using both standard risk factors and ten previously identified SNVs. The addition of genetic information modestly changed the AUC from 58% to 61.8%, a result that was not felt to be clinically significant. Despite this, 32.5% of patients were in a higher quintile of breast cancer risk when genetic information was included, and 20.4% were in a lower quintile of risk. Whether such information has clinical utility is unclear.[25,28]

More limited data are available regarding ovarian cancer risk. Three GWAS involving staged analysis of more than 10,000 cases and 13,000 controls have been carried out for ovarian cancer.[29-31] As in other GWAS, the ORs are modest, generally about 1.2 or weaker but implicate a number of genes with plausible biological ties to ovarian cancer, such as BABAM1, whose protein complexes with and may regulate BRCA1, and TIRAPR, which codes for a poly (ADP-ribose) polymerase, molecules that may be important in BRCA1/BRCA2-deficient cells.

Polygenic risk scores for breast and ovarian cancer

The collective influence of many genetic variants has more recently been evaluated using an aggregate score. In 2015, a polygenic risk score (PRS) comprising all of the known breast cancer risk genetic variants or SNVs was estimated in women of European ancestry using 41 studies in the Breast Cancer Association Consortium (BCAC), including more than 33,000 breast cancer cases and 33,000 controls.[32] This early attempt at estimating a PRS for breast cancer included 77 SNVs, which collectively conferred lifetime risks of developing breast cancer by age 80 years of 3.5% and 29% for women in the lowest and highest 1% of the PRS, respectively.[32] Since then, PRSs incorporating additional genetic variants and examining other breast cancer–related outcomes including tumor and pathological characteristics, mode of detection, and contralateral breast cancer (CBC) have been estimated.[33-40] In 2019, the PRS with the highest discriminatory ability to date was developed and prospectively validated in the largest GWAS datasets available (79 studies in BCAC and more than 190,000 women in the U.K. Biobank), which incorporates information on 313 genetic variants and is optimized for ER-positive and ER-negative breast cancer.[39] Compared with women in the middle quintile, those in the highest 1% of PRS313 had 4.04-, 4.37-, and 2.78-fold risks of developing breast cancer overall, ER-positive disease, and ER-negative disease, respectively.[39] Lifetime absolute risk of breast cancer by age 80 years for women in the lowest and highest 1% of PRS313 ranged from 2% to 31% for ER-positive breast cancer, while for ER-negative disease, the absolute risks ranged from 0.55% to 4%.[39]

Common genomic variants associated with the development of a first primary breast cancer are also associated with the development of CBC.[40] Women in the highest quartile of the PRS had a 1.6-fold increased risk of developing CBC compared with the lowest quartile.[40] Moreover, PRSs of breast and ovarian cancers have been assessed in women who are carriers of BRCA1 and BRCA2 pathogenic variants, and have been found to be predictive of cancer risk in these women, supporting the hypothesis of a shared polygenic component of cancer risk between the general population and variant carriers.[36] The PRS for ER-negative disease had the strongest association with breast cancer risk in BRCA1 variant carriers, while the strongest association in BRCA2 variant carriers was seen for the overall breast cancer PRS. BRCA1 variant carriers had cumulative lifetime risks of 56% and 75% of developing breast cancer at the 10th and 90th percentile of the PRS, respectively. The ovarian cancer PRS was strongly associated with risk for both BRCA1 and BRCA2 variant carriers. For BRCA2 variant carriers, the ovarian cancer risk was 6% and 19% by age 80 years for those at the 10th and 90th percentile of PRS, respectively. The authors noted that the incorporation of the PRS into risk prediction models may better inform decisions on cancer risk management for this population.[36]

Several studies have examined the extent to which clinical breast cancer risk prediction models can be improved by including information on known susceptibility SNVs, and reporting improved discriminatory accuracy after inclusion of the PRS.[41-46] For example, in a study combining PRS77 with clinical models, the AUC for predicting breast cancer before age 50 years improved by more than 20%.[42] Clinical trials, including WISDOM and MyPeBs, are in progress to study the potential clinical utility of the PRS for making screening decisions and understanding outcomes.[47] Because PRSs have been largely developed and validated in populations of European ancestry, the utility and prediction accuracy of these PRSs in non-European populations is unknown.

A large study examined whether known reproductive and lifestyle risk factors interact with PRSs to increase breast cancer risk and did not find a multiplicative interaction with established risk factors.[48]

Whole-Genome and Whole-Exome Sequencing

In addition to GWAS interrogating common genetic variants, sequencing-based studies involving whole-genome or whole-exome sequencing [49] are also identifying genes associated with breast cancer, such as XRCC2, a rare, moderate-penetrance breast cancer susceptibility gene.[50] (Refer to the Clinical Sequencing section in the PDQ summary on Cancer Genetics Overview for more information about whole-exome sequencing.)

References
  1. Pharoah PD, Antoniou A, Bobrow M, et al.: Polygenic susceptibility to breast cancer and implications for prevention. Nat Genet 31 (1): 33-6, 2002. [PUBMED Abstract]
  2. Breast Cancer Association Consortium: Commonly studied single-nucleotide polymorphisms and breast cancer: results from the Breast Cancer Association Consortium. J Natl Cancer Inst 98 (19): 1382-96, 2006. [PUBMED Abstract]
  3. Dunning AM, Healey CS, Pharoah PD, et al.: A systematic review of genetic polymorphisms and breast cancer risk. Cancer Epidemiol Biomarkers Prev 8 (10): 843-54, 1999. [PUBMED Abstract]
  4. Thorisson GA, Smith AV, Krishnan L, et al.: The International HapMap Project Web site. Genome Res 15 (11): 1592-3, 2005. [PUBMED Abstract]
  5. Clarke L, Zheng-Bradley X, Smith R, et al.: The 1000 Genomes Project: data management and community access. Nat Methods 9 (5): 459-62, 2012. [PUBMED Abstract]
  6. Evans DM, Cardon LR: Genome-wide association: a promising start to a long race. Trends Genet 22 (7): 350-4, 2006. [PUBMED Abstract]
  7. Cardon LR: Genetics. Delivering new disease genes. Science 314 (5804): 1403-5, 2006. [PUBMED Abstract]
  8. Chanock SJ, Manolio T, Boehnke M, et al.: Replicating genotype-phenotype associations. Nature 447 (7145): 655-60, 2007. [PUBMED Abstract]
  9. Wellcome Trust Case Control Consortium: Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447 (7145): 661-78, 2007. [PUBMED Abstract]
  10. Easton DF, Pooley KA, Dunning AM, et al.: Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 447 (7148): 1087-93, 2007. [PUBMED Abstract]
  11. Stacey SN, Manolescu A, Sulem P, et al.: Common variants on chromosomes 2q35 and 16q12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 39 (7): 865-9, 2007. [PUBMED Abstract]
  12. Hunter DJ, Kraft P, Jacobs KB, et al.: A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat Genet 39 (7): 870-4, 2007. [PUBMED Abstract]
  13. Turnbull C, Ahmed S, Morrison J, et al.: Genome-wide association study identifies five new breast cancer susceptibility loci. Nat Genet 42 (6): 504-7, 2010. [PUBMED Abstract]
  14. Gold B, Kirchhoff T, Stefanov S, et al.: Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33. Proc Natl Acad Sci U S A 105 (11): 4340-5, 2008. [PUBMED Abstract]
  15. Zheng W, Long J, Gao YT, et al.: Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat Genet 41 (3): 324-8, 2009. [PUBMED Abstract]
  16. Kibriya MG, Jasmine F, Argos M, et al.: A pilot genome-wide association study of early-onset breast cancer. Breast Cancer Res Treat 114 (3): 463-77, 2009. [PUBMED Abstract]
  17. Murabito JM, Rosenberg CL, Finger D, et al.: A genome-wide association study of breast and prostate cancer in the NHLBI's Framingham Heart Study. BMC Med Genet 8 (Suppl 1): S6, 2007. [PUBMED Abstract]
  18. Stacey SN, Manolescu A, Sulem P, et al.: Common variants on chromosome 5p12 confer susceptibility to estrogen receptor-positive breast cancer. Nat Genet 40 (6): 703-6, 2008. [PUBMED Abstract]
  19. Ahmed S, Thomas G, Ghoussaini M, et al.: Newly discovered breast cancer susceptibility loci on 3p24 and 17q23.2. Nat Genet 41 (5): 585-90, 2009. [PUBMED Abstract]
  20. Reeves GK, Travis RC, Green J, et al.: Incidence of breast cancer and its subtypes in relation to individual and multiple low-penetrance genetic susceptibility loci. JAMA 304 (4): 426-34, 2010. [PUBMED Abstract]
  21. Haiman CA, Chen GK, Vachon CM, et al.: A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor-negative breast cancer. Nat Genet 43 (12): 1210-4, 2011. [PUBMED Abstract]
  22. Stevens KN, Fredericksen Z, Vachon CM, et al.: 19p13.1 is a triple-negative-specific breast cancer susceptibility locus. Cancer Res 72 (7): 1795-803, 2012. [PUBMED Abstract]
  23. Campa D, Kaaks R, Le Marchand L, et al.: Interactions between genetic variants and breast cancer risk factors in the breast and prostate cancer cohort consortium. J Natl Cancer Inst 103 (16): 1252-63, 2011. [PUBMED Abstract]
  24. Milne RL, Gaudet MM, Spurdle AB, et al.: Assessing interactions between the associations of common genetic susceptibility variants, reproductive history and body mass index with breast cancer risk in the breast cancer association consortium: a combined case-control study. Breast Cancer Res 12 (6): R110, 2010. [PUBMED Abstract]
  25. Pharoah PD, Antoniou AC, Easton DF, et al.: Polygenes, risk prediction, and targeted prevention of breast cancer. N Engl J Med 358 (26): 2796-803, 2008. [PUBMED Abstract]
  26. Gail MH: Discriminatory accuracy from single-nucleotide polymorphisms in models to predict breast cancer risk. J Natl Cancer Inst 100 (14): 1037-41, 2008. [PUBMED Abstract]
  27. Gail MH: Value of adding single-nucleotide polymorphism genotypes to a breast cancer risk model. J Natl Cancer Inst 101 (13): 959-63, 2009. [PUBMED Abstract]
  28. Wacholder S, Hartge P, Prentice R, et al.: Performance of common genetic variants in breast-cancer risk models. N Engl J Med 362 (11): 986-93, 2010. [PUBMED Abstract]
  29. Song H, Ramus SJ, Tyrer J, et al.: A genome-wide association study identifies a new ovarian cancer susceptibility locus on 9p22.2. Nat Genet 41 (9): 996-1000, 2009. [PUBMED Abstract]
  30. Goode EL, Chenevix-Trench G, Song H, et al.: A genome-wide association study identifies susceptibility loci for ovarian cancer at 2q31 and 8q24. Nat Genet 42 (10): 874-9, 2010. [PUBMED Abstract]
  31. Bolton KL, Tyrer J, Song H, et al.: Common variants at 19p13 are associated with susceptibility to ovarian cancer. Nat Genet 42 (10): 880-4, 2010. [PUBMED Abstract]
  32. Mavaddat N, Pharoah PD, Michailidou K, et al.: Prediction of breast cancer risk based on profiling with common genetic variants. J Natl Cancer Inst 107 (5): , 2015. [PUBMED Abstract]
  33. Curtit E, Pivot X, Henriques J, et al.: Assessment of the prognostic role of a 94-single nucleotide polymorphisms risk score in early breast cancer in the SIGNAL/PHARE prospective cohort: no correlation with clinico-pathological characteristics and outcomes. Breast Cancer Res 19 (1): 98, 2017. [PUBMED Abstract]
  34. Cuzick J, Brentnall AR, Segal C, et al.: Impact of a Panel of 88 Single Nucleotide Polymorphisms on the Risk of Breast Cancer in High-Risk Women: Results From Two Randomized Tamoxifen Prevention Trials. J Clin Oncol 35 (7): 743-750, 2017. [PUBMED Abstract]
  35. Khera AV, Chaffin M, Aragam KG, et al.: Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet 50 (9): 1219-1224, 2018. [PUBMED Abstract]
  36. Kuchenbaecker KB, McGuffog L, Barrowdale D, et al.: Evaluation of Polygenic Risk Scores for Breast and Ovarian Cancer Risk Prediction in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst 109 (7): , 2017. [PUBMED Abstract]
  37. Li H, Feng B, Miron A, et al.: Breast cancer risk prediction using a polygenic risk score in the familial setting: a prospective study from the Breast Cancer Family Registry and kConFab. Genet Med 19 (1): 30-35, 2017. [PUBMED Abstract]
  38. Li J, Ugalde-Morales E, Wen WX, et al.: Differential Burden of Rare and Common Variants on Tumor Characteristics, Survival, and Mode of Detection in Breast Cancer. Cancer Res 78 (21): 6329-6338, 2018. [PUBMED Abstract]
  39. Mavaddat N, Michailidou K, Dennis J, et al.: Polygenic Risk Scores for Prediction of Breast Cancer and Breast Cancer Subtypes. Am J Hum Genet 104 (1): 21-34, 2019. [PUBMED Abstract]
  40. Robson ME, Reiner AS, Brooks JD, et al.: Association of Common Genetic Variants With Contralateral Breast Cancer Risk in the WECARE Study. J Natl Cancer Inst 109 (10): , 2017. [PUBMED Abstract]
  41. Allman R, Dite GS, Hopper JL, et al.: SNPs and breast cancer risk prediction for African American and Hispanic women. Breast Cancer Res Treat 154 (3): 583-9, 2015. [PUBMED Abstract]
  42. Dite GS, MacInnis RJ, Bickerstaffe A, et al.: Breast Cancer Risk Prediction Using Clinical Models and 77 Independent Risk-Associated SNPs for Women Aged Under 50 Years: Australian Breast Cancer Family Registry. Cancer Epidemiol Biomarkers Prev 25 (2): 359-65, 2016. [PUBMED Abstract]
  43. Shieh Y, Hu D, Ma L, et al.: Breast cancer risk prediction using a clinical risk model and polygenic risk score. Breast Cancer Res Treat 159 (3): 513-25, 2016. [PUBMED Abstract]
  44. Starlard-Davenport A, Allman R, Dite GS, et al.: Validation of a genetic risk score for Arkansas women of color. PLoS One 13 (10): e0204834, 2018. [PUBMED Abstract]
  45. van Veen EM, Brentnall AR, Byers H, et al.: Use of Single-Nucleotide Polymorphisms and Mammographic Density Plus Classic Risk Factors for Breast Cancer Risk Prediction. JAMA Oncol 4 (4): 476-482, 2018. [PUBMED Abstract]
  46. Zhang X, Rice M, Tworoger SS, et al.: Addition of a polygenic risk score, mammographic density, and endogenous hormones to existing breast cancer risk prediction models: A nested case-control study. PLoS Med 15 (9): e1002644, 2018. [PUBMED Abstract]
  47. Esserman LJ; WISDOM Study and Athena Investigators: The WISDOM Study: breaking the deadlock in the breast cancer screening debate. NPJ Breast Cancer 3: 34, 2017. [PUBMED Abstract]
  48. Rudolph A, Song M, Brook MN, et al.: Joint associations of a polygenic risk score and environmental risk factors for breast cancer in the Breast Cancer Association Consortium. Int J Epidemiol 47 (2): 526-536, 2018. [PUBMED Abstract]
  49. Shendure J: Next-generation human genetics. Genome Biol 12 (9): 408, 2011. [PUBMED Abstract]
  50. Park DJ, Lesueur F, Nguyen-Dumont T, et al.: Rare mutations in XRCC2 increase the risk of breast cancer. Am J Hum Genet 90 (4): 734-9, 2012. [PUBMED Abstract]

Clinical Management of Carriers of BRCA Pathogenic Variants

Increasing data are available on the outcomes of interventions to reduce risk in people with a genetic susceptibility to breast cancer or ovarian cancer.[1-7] As outlined in other sections of this summary, uncertainty is often considerable regarding the level of cancer risk associated with a positive family history or genetic test. In this setting, personal preferences are likely to be an important factor in patients’ decisions about risk reduction strategies.

Screening and Prevention Strategies

Breast cancer

Screening/surveillance

Refer to the PDQ summary on Breast Cancer Screening for information on screening in the general population, and to the PDQ summary Levels of Evidence for Cancer Genetics Studies for information on levels of evidence related to screening and prevention.

Breast self-examination

In the general population, evidence for the value of breast self-examination (BSE) is limited. Preliminary results have been reported from a randomized study of BSE being conducted in Shanghai, China.[8] At 5 years, no reduction in breast cancer mortality was seen in the BSE group compared with the control group of women, nor was a substantive stage shift seen in breast cancers that were diagnosed. (Refer to the PDQ summary on Breast Cancer Screening for more information.)

Little direct prospective evidence exists regarding BSE in individuals with an increased risk of breast cancer. In the Canadian National Breast Screening Study, women with first-degree relatives (FDRs) with breast cancer had statistically significantly higher BSE competency scores than those without a family history. In a study of 251 high-risk women at a referral center, five breast cancers were detected by self-examination less than a year after a previous screen (as compared with one cancer detected by clinician exam and 11 cancers detected as a result of mammography). Women in the cohort were instructed in self-examination, but it is not stated whether the interval cancers were detected as a result of planned self-examination or incidental discovery of breast masses.[9] In another series of carriers of BRCA1/BRCA2 pathogenic variants, four of nine incident cancers were diagnosed as palpable masses after a reportedly normal mammogram, further suggesting the potential value of self-examination.[10] A task force convened by the Cancer Genetics Studies Consortium has recommended “monthly self-examination beginning early in adult life (e.g., by age 18–21 y) to establish a regular habit and allow familiarity with the normal characteristics of breast tissue. Education and instruction in self-examination are recommended.”[11]

Level of evidence: 5

Clinical breast examination

Few prospective data exist regarding clinical breast examination (CBE).

The Cancer Genetics Studies Consortium task force concluded, “As with self-examination, the contribution of clinical examination may be particularly important for women at inherited risk of early breast cancer.” They recommended that female carriers of a BRCA1 or BRCA2 high-risk pathogenic variant undergo annual or semiannual clinical examinations beginning at age 25 to 35 years.[11]

Level of evidence: 5

Mammography

In the general population, strong evidence suggests that regular mammography screening of women aged 50 to 59 years leads to a 25% to 30% reduction in breast cancer mortality. (Refer to the PDQ summary on Breast Cancer Screening for more information.) For women who begin mammographic screening at age 40 to 49 years, a 17% reduction in breast cancer mortality is seen, which occurs 15 years after the start of screening.[12] Observational data from a cohort study of more than 28,000 women suggest that the sensitivity of mammography is lower for young women. In this study, the sensitivity was lowest for younger women (aged 30–49 y) who had an FDR with breast cancer. For these women, mammography detected 69% of breast cancers diagnosed within 13 months of the first screening mammography. By contrast, sensitivity for women younger than 50 years without a family history was 88% (P = .08). For women aged 50 years and older, sensitivity was 93% at 13 months and did not vary by family history.[13] Preliminary data suggest that mammography sensitivity is lower in BRCA1 and BRCA2 carriers than in noncarriers.[10] Subsequent observational studies have found that the positive predictive value (PPV) of mammography increases with age and is highest among older women and among women with a family history of breast cancer.[14] Higher PPVs may be due to increased breast cancer incidence, higher sensitivity, and/or higher specificity.[15] One study found an association between the presence of pushing margins and false-negative mammograms in 28 women, 26 of whom had a BRCA1 pathogenic variant and two of whom had a BRCA2 pathogenic variant. Pushing margins, a mammographic characteristic of medullary histology, are associated with an absence of fibrotic reaction.[16] In addition, rapid tumor doubling times may lead to tumors presenting shortly after an apparently normal study. In one study, mean tumor doubling time in BRCA1/BRCA2 carriers was 45 days, compared with 84 days in noncarriers.[17] Another study that evaluated mammographic breast density in women with BRCA pathogenic variants found no association between pathogenic variant status and mammographic density; however, in both carriers and noncarriers, increased breast density was associated with increased breast cancer risk.[18]

The randomized Canadian National Breast Screening Study-2 compared annual CBE plus mammography to CBE alone in women aged 50 to 59 years from the general population. Both groups were given instruction in BSE.[19] Although mammography detected smaller primary invasive tumors, more invasive cancers, and more ductal carcinoma in situ (DCIS) than CBE, the breast cancer mortality rates in the CBE-plus-mammography group and the CBE-alone group were nearly identical, and compared favorably with other breast cancer screening trials. After a mean follow-up of 13 years (range, 11.3–16.0 y), the cumulative breast cancer mortality ratio was 1.02 (95% confidence interval [CI], 0.78–1.33). One possible explanation of this finding was the careful training and supervision of the health professionals performing CBE.

Digital mammography refers to the use of a digital detector to find and record x-ray images. This technology improves contrast resolution [20] and has been proposed as a potential strategy for improving the sensitivity of mammography. A screening study comparing digital with routine mammography in 6,736 examinations of women aged 40 years and older found no difference in cancer detection rates;[21] however, digital mammography resulted in fewer recalls. In another study (ACRIN-6652) comparing digital mammography to plain-film mammography in 42,760 women, the overall diagnostic accuracy of the two techniques was similar.[22] When receiver operating characteristic curves were compared, digital mammography was more accurate in women younger than 50 years, in women with radiographically dense breasts, and in premenopausal or perimenopausal women.

In a prospective study of 251 individuals with BRCA pathogenic variants who received uniform recommendations regarding screening and risk-reducing surgery, annual mammography detected breast cancer in six women at a mean of 20.2 months after receipt of BRCA results.[9] The Cancer Genetics Studies Consortium task force has recommended that female carriers of a BRCA1 or BRCA2 high-risk pathogenic variant have, “annual mammography, beginning at age 25 to 35 years. Mammograms should be done at a consistent location when possible, with prior films available for comparison.”[11] Data from prospective studies on the relative benefits and risks of screening with an ionizing radiation tool versus CBE or other nonionizing radiation tools would be useful.[23-25]

Certain observations raise concerns that carriers of BRCA pathogenic variants may be more prone to radiation-induced breast cancer than women without pathogenic variants. The BRCA1 and BRCA2 proteins are known to be important in cellular mechanisms of DNA damage repair, including those involved in repairing radiation-induced damage. Some studies have suggested intermediate radiation sensitivity in cells that are heterozygous for a BRCA variant, but this is not consistent and varies by experimental system and endpoint.

Three studies have failed to find convincing evidence of an association between ionizing radiation exposure and increased breast cancer risk in carriers of BRCA1 and BRCA2 pathogenic variants.[26-28] In contrast, two large international studies found evidence of an increased breast cancer risk due to chest x-rays [29] or estimates of total exposure to diagnostic radiation.[30] A large, international, case-control study of 1,601 carriers of pathogenic variants described an increased risk of breast cancer (hazard ratio [HR], 1.54) among women who were ever exposed to chest x-rays, with risk being highest in women aged 40 years and younger, born after 1949, and exposed to x-rays only before age 20 years.[29] Some of the subjects in this study were also included in a larger, more comprehensive analysis of carriers of pathogenic variants from three European centers.[30] In this study of 1,993 carriers of BRCA1 and BRCA2 pathogenic variants from the United Kingdom, France, and the Netherlands, age-specific total diagnostic radiation exposure (e.g., chest x-rays, mammography, fluoroscopy, and computed tomography) estimates were derived from self-reported questionnaires. Women who were exposed to radiation before age 30 years had an increased risk (HR, 1.90; 95% CI, 1.20–3.00) when compared with those who were never exposed. This risk was primarily driven by nonmammographic radiation exposure in women younger than 20 years (HR, 1.62; 95% CI, 1.02–2.58). Subsequently, a prospective study of 1,844 BRCA1 carriers and 502 BRCA2 carriers without a breast cancer diagnosis at study entry, with an average follow-up time of 5.3 years, observed no significant association between prior mammography exposure and breast cancer risk.[28] Additional subgroup analyses in women younger than 30 years demonstrated no association with breast cancer risk.

With the routine use of magnetic resonance imaging (MRI) in carriers of BRCA1 and BRCA2 pathogenic variants, any potential benefit of mammographic screening must be carefully weighed against potential risks, particularly in young women.[31] One study has suggested that the most cost-effective screening strategy in carriers of BRCA1 and BRCA2 pathogenic variants may be annual MRI beginning at age 25 years, with alternating MRI and digital mammography (so that each test is done annually but screening occurs every 6 months) beginning at age 30 years.[32] The National Comprehensive Cancer Network (NCCN) currently recommends annual breast MRI screening with contrast (or mammogram with consideration of tomosynthesis, only if MRI is unavailable) between ages 25 and 29 years and annual mammogram (with consideration of tomosynthesis and breast MRI screening with contrast) between ages 30 and 75 years.[33]

Magnetic resonance imaging

Because of the relative insensitivity of mammography in women with an inherited risk of breast cancer, a number of screening modalities have been proposed and investigated in high-risk women, including carriers of BRCA pathogenic variants. Many studies have described the experience with breast MRI screening in women at risk of breast cancer, including descriptions of relatively large multi-institutional trials.[34-42]

Despite some limitations, these studies consistently demonstrate that breast MRI is more sensitive than either mammography or ultrasound for the detection of hereditary breast cancer. The results of six large studies are presented in Table 11, Summary of MRI Screening Studies in Women at Hereditary Risk of Breast Cancer.[34,36,37,40,43,44] Most cancers in these programs were screen detected, with only 6% of cancers presenting in the interval between screenings. The sensitivity of MRI (as defined by the study methodology) ranged from 71% to 100%. Of the combined studies, 77% of cancers were identified by MRI, and 42% were identified by mammography.

Concerns have been raised about the reduced specificity of MRI compared with other screening modalities. In one study, after the initial MRI screen, 16.5% of patients were recalled for further evaluation and an additional 7.6% of patients were recommended to undergo a short-interval follow-up examination at 6 months.[37] These rates declined significantly during later screening rounds, with fewer than 10% of the subjects recalled for more detailed MRI and fewer than 3% recommended to have short interval follow-up. In a second study, Magnetic Resonance Imaging for Breast Screening (MARIBS), the recall rate for additional evaluation was 10.7% per year.[36] The benign biopsy rates in the first study were 11% at first round, 6.6% at second round, and 4.7% at third round.[37] In the MARIBS study, the aggregate surgical biopsy rate was 9 per 1,000 screening episodes, though this may underestimate the burden because follow-up ultrasounds, core-needle biopsies, and fine-needle aspirations have not been included in the numerator of the MARIBS calculation.[36] The PPV of MRI has been calculated differently in the various series and fluctuates somewhat, depending on whether all abnormal examinations or only the examinations that result in a biopsy are counted in the denominator. Generally, the PPV of a recommendation for tissue sampling (as opposed to further investigation) is in the range of 50% in most series.

These trials appear to establish that MRI is superior to mammography in the detection of hereditary breast cancer; women who participated in these trials, which included annual MRI, were less likely to have a cancer missed by screening.[45] However, mammography may identify some cancers, particularly DCIS, that are not identified by MRI.[46]

Regarding downstaging, one screening study demonstrated that patients at risk of hereditary breast cancer were more likely to be diagnosed with small tumors and node-negative disease than were women in two nonrandomized control groups.[34] A multicenter randomized trial compared annual breast MRI, annual CBE, and biannual mammogram with annual mammogram and CBE among women who tested negative for BRCA1/BRCA2 pathogenic variants but with a lifetime familial risk of breast cancer of 20% or higher. Invasive cancers in the MRI arm were smaller, had fewer positive nodes, and were diagnosed at lower stages than those in the mammogram-alone arm.[47]

Despite the apparent sensitivity of MRI screening, some women in MRI-based programs will develop life-threatening breast cancer. In a prospective study of 51 carriers of BRCA1 pathogenic variants and 41 carriers of BRCA2 pathogenic variants screened with yearly mammograms and MRIs (of whom 80 had risk-reducing oophorectomy), 11 breast cancers (9 invasive and 2 DCIS) were detected. Six cancers were first detected on MRI; three were first detected by mammogram; and two were interval cancers. All breast cancers occurred in carriers of BRCA1 pathogenic variants, suggesting a continued high risk of BRCA1-related breast cancer after oophorectomy in the short term. These results suggest that surveillance and prevention strategies may have differing outcomes in carriers of BRCA1 and BRCA2 pathogenic variants.[41]

A publication combining results from three large studies (MARIBS, a Canadian study, and a Dutch MRI screening study) demonstrated that when MRI was added to mammography, 80% of cancers detected in carriers of BRCA2 pathogenic variants were either DCIS or invasive cancers smaller than 1 cm. In carriers of BRCA1 pathogenic variants, 49% of cancers were DCIS or small invasive cancers. In addition, the authors predicted mortality benefits with the addition of MRI for both carriers of BRCA1 and BRCA2 pathogenic variants. The model predicted breast cancer mortality reductions of 42% to 47% for mammography, 48% to 61% for MRI, and 50% to 62% for combined screening.[48] An additional study examining carriers of BRCA1/BRCA2 pathogenic variants undergoing MRI between 1997 and 2006 has demonstrated that 97% of incident cancers were stage 0 or stage I.[49] A 2015 Dutch case-control study further evaluated 2,308 high-risk patients, including 706 women with known BRCA pathogenic variants, who were screened with mammogram and compared them with those who also had MRI.[50] Of the patients screened, 93 patients were detected to have 97 cancers, 33 patients had a BRCA1 pathogenic variant, and 18 patients had a BRCA2 pathogenic variant. With a median follow-up of 9 years, metastases-free survival was improved in the MRI-screened cohort (90% vs. 77%), but it did not reach statistical significance in the BRCA1 and BRCA2 subset because of a very small sample size. All MRI-screened patients were more likely to be node-negative and receive less chemotherapy. The American Cancer Society and NCCN have recommended the use of annual MRI screening for women at hereditary risk of breast cancer.[33,51]

An additional question regarding the timing of mammography and MRI is whether they should be done simultaneously or in an alternating fashion (so that while each test is done annually, screening occurs every 6 months). One study has suggested that the most cost-effective screening strategy in carriers of BRCA1 and BRCA2 pathogenic variants may be annual MRI beginning at age 25 years, with alternating MRI and digital mammography beginning at age 30 years.[32]

A study of women who had undergone risk-reducing mastectomy reported a low yield from annual MRI screening based on a retrospective review of 159 women, 58% of whom had pathogenic BRCA variants, over an 8-year period. MRI was negative in 98% of patients and was associated with a false-positive rate of 90%. The yield of surveillance MRI was very low in this cohort, bringing into question the added value of routine surveillance MRI in this population.[52]

In summary, evidence strongly supports the integral role of breast MRI in breast cancer surveillance for carriers of BRCA1/BRCA2 pathogenic variants.

Table 11. Summary of Magnetic Resonance Imaging (MRI) Screening Studies in Women at Hereditary Risk of Breast Cancer
Series Rijnsburger [42] Warner [37] MARIBS [36] Kuhl [40] Weinstein [43] Sardanelli [44] Totals
aBased on the first 1,909 women screened.[34]
bIncludes patients with invasive cancer only and patients with both invasive and in situ cancers.
cIncludes only 75 cancers detected in women who underwent both mammographic and MRI screening.
dRestricted to studies in which ultrasound was performed.
N Patients Overall 2,157 236 649 687 609 501 4,839
BRCA1/BRCA2 Carriers 594 236 120 65 44 330 1,389
N Screening Episodes 6,253 457 1,881 1,679   1,592 11,862
N Cancers Baseline 22a 13 20 10 0 0 65
Subsequent 97 9 15 17 18 52 208
Invasiveb 78 16 29 8 11 44 186
In situ 19 9 6 9 7 8 58
Annual Incidence 10.4/1,000   19/1,000        
Detected at Planned Screening 78 21 33 27 18 49 226 (83%)
N Detected by Each Modality Mammography 31c 8 14 9 7 25 94 (42%)
MRI 51c 17 27 25 12 42 174 (77%)
Ultrasoundd   7   10 3 26 46 (41%)
Follow-up Median of 4.9 y Minimum of 1 y 2–7 y Median of 29.09 mo 2 y 3 y  

Level of evidence: 3

Ultrasound

Several studies have reported instances of breast cancer detected by ultrasound that were missed by mammography, as discussed in one review.[53] In a pilot study of ultrasound as an adjunct to mammography in 149 women with moderately increased risk based on family history, one cancer was detected, based on ultrasound findings. Nine other biopsies of benign lesions were performed. One was based on abnormalities on both mammography and ultrasound, and the remaining eight were based on abnormalities on ultrasound alone.[53] A large study of 2,809 women with dense breast tissue (ACRIN-6666) demonstrated that ultrasound increased the detection rate due to breast cancer screening from 7.6 per 1,000 with mammography alone to 11.8 per 1,000 for combined mammography and ultrasound.[54] However, ultrasound screening increases false-positive rates and appears to have a limited benefit in combination with MRI. In a multicenter study of 171 women (92% of whom were carriers of BRCA1/BRCA2 pathogenic variants) undergoing simultaneous mammography, MRI, and ultrasound, no cancers were detected by ultrasound alone.[38] Uncertainties about ultrasound include the effect of screening on mortality, the rate and outcome of false-positive results, and access to experienced breast ultrasonographers.

Level of evidence: None assigned

Other screening modalities

A number of other techniques are under active investigation, including tomosynthesis, contrast-enhanced mammography, thermography, and radionuclide scanning. Additional evidence is needed before these techniques can be incorporated into clinical practice.

Level of evidence: None assigned

Risk-reducing surgeries
Risk-reducing mastectomy

Risk-reducing mastectomy (RRM) is a management option for patients who are considered to be at high risk of developing breast cancer. The Society of Surgical Oncology has endorsed RRM as an option for women with BRCA1/BRCA2 pathogenic variants or strong family histories of breast cancer.[55] Historically, a total or simple mastectomy has been performed, which includes removal of all of the breast tissue, including the nipple and areolar complex (NAC). If the patient is interested, reconstruction can be performed simultaneously with the ablative portion of the procedure. Options for reconstruction include tissue expander and implant-based reconstructions or autologous reconstructions, in which the patient’s own tissue is used to reconstruct the breast. A number of different tissues can be used to reconstruct the breast, including flaps based on the latissimus dorsi muscle, the transverse rectus abdominis muscle, or the gluteus muscle. Muscle-sparing techniques such as the deep inferior epigastric perforator flap can also be used, but these require advanced microvascular techniques. In the interest of improved cosmetic outcomes, skin-sparing techniques have been utilized in which the entire breast is removed with the NAC, but the entire skin envelope of the breast is preserved. In a further refinement, nipple-sparing techniques have been developed in which all of the breast skin and the nipple are preserved while the underlying glandular tissue is removed.

Risk-reducing mastectomy in unaffected women

Because there are no randomized, prospective trials of RRM versus observation, data are limited to cohort and case-control studies. The available data demonstrate that RRM does decrease breast cancer incidence in high-risk patients,[56-58] but overall survival (OS) correlates more closely with the overall risk from the primary incidence of breast cancer. Several studies have analyzed the impact of RRM on breast cancer risk and mortality. In one retrospective cohort study of 214 women considered to be at hereditary risk by virtue of a family history suggesting an autosomal dominant predisposition, three women were diagnosed with breast cancer after bilateral RRM, with a median follow-up of 14 years.[58] Because 37.4 cancers were expected, the calculated risk reduction was 92.0% (95% CI, 76.6%–98.3%). In a follow-up subset analysis, 176 of the 214 high-risk women in this cohort study underwent genetic testing for pathogenic variants in BRCA1 and BRCA2. Pathogenic variants were identified in 18 women, none of whom developed breast cancer after a median follow-up of 13.4 years.[56] Two of the three women diagnosed with breast cancer after RRM were tested, and neither carried a pathogenic variant. The calculated risk reduction among carriers of pathogenic variants was 89.5% to 100.0% (95% CI, 41.4%–100.0%), depending on the assumptions made about the expected numbers of cancers among carriers of pathogenic variants and the status of the untested woman who developed cancer despite mastectomy. The result of this retrospective cohort study has been supported by a prospective analysis of 76 carriers of pathogenic variants who underwent RRM and were monitored prospectively for a mean of 2.9 years. No breast cancers were observed in these women, whereas eight were identified in women who underwent regular surveillance (HR for breast cancer after RRM, 0.00 [95% CI, 0.00–0.36]).[57]

The Prevention and Observation of Surgical Endpoints study group also estimated the degree of breast cancer risk reduction after RRM in carriers of BRCA1/BRCA2 pathogenic variants. The rate of breast cancer in 105 carriers of pathogenic variants who underwent bilateral RRM was compared with that of 378 carriers who did not choose surgery. Bilateral mastectomy reduced the risk of breast cancer by approximately 90% after a mean follow-up of 6.4 years.[3]

Theoretical models have also been utilized to assess the role of RRM in women with pathogenic variants in BRCA1 and BRCA2. Assuming risk reduction in the range of 90%, one model suggests that for a group of women, aged 30 years, with BRCA1 or BRCA2 pathogenic variants, RRM would result in an average increased life expectancy of 2.9 to 5.3 years.[59] A computer-simulated survival analysis using a Monte Carlo model included breast MRI, mammography, RRM, and risk-reducing salpingo-oophorectomy (RRSO) and examined the impact of each intervention separately on carriers of BRCA1 and BRCA2 pathogenic variants.[5] The most effective strategy was found to be RRSO at age 40 years and RRM at age 25 years, with survival at age 70 years approaching that of the general population. However, delaying mastectomy until age 40 years, or substituting RRM with screening with breast MRI and mammography, had little impact on survival estimates. For example, replacing RRM with MRI-based screening in women with RRSO at age 40 years led to a 3% to 5% decrement in survival compared with RRM at age 25 years.[60] As with any models, numerous assumptions cause uncertainty; however, these studies provide additional information for women and their providers who are making these difficult decisions.

Another study of at-risk women showed a 70% time–trade-off value for RRM, indicating that participants were willing to sacrifice 30% of life expectancy to avoid RRM.[61] A cost-effectiveness analysis study of RRM has also been performed. The investigators concluded that, compared with surveillance, risk-reducing surgery (mastectomy and oophorectomy) is cost-effective with regard to years of life saved, but not for improved quality of life.[62] While these data are interesting and may be useful for public policy decisions, they cannot be individualized for clinical care because they include assumptions that cannot be fully tested.

Level of evidence: 3ai

Contralateral risk-reducing mastectomy in affected women

If RRM is effective in lowering breast cancer risk in unaffected women, what is its role in women with unilateral breast cancer? This question often arises in discussions about surgical options with women who have unilateral breast cancer and hereditary risks. This section addresses the role of contralateral risk-reducing mastectomy (CRRM) in women being treated with mastectomy and will not discuss breast conservation therapy. Multiple studies have shown an increase in the rate of CRRM in women with unilateral breast cancer.[63,64] When the appropriateness of CRRM is being assessed for women with unilateral breast cancer, the first task is to determine the risk of contralateral breast cancer (CBC).

In the general population, current estimates of CBC risk after breast cancer treatment are approximately 0.3% per year and are declining.[65] In carriers of BRCA pathogenic variants with a diagnosis of breast cancer, the risk of a second, unrelated breast cancer is related to age at initial diagnosis, biology, and systemic therapies used, but is clearly higher than that in the general population.[66] (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section in the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for more information about the risk of CBC in this population.) In carriers of BRCA pathogenic variants whose first cancer has an excellent prognosis, estimating the risk of a second, unrelated breast cancer event is important for informing their decision to undergo risk-reducing surgery and has been described in this setting to improve survival.[67] The timing of genetic testing and knowledge of BRCA pathogenic variant status may influence surgical decision making, may prevent subsequent surgeries, and may influence follow-up care. Therefore, for individuals at increased risk of carrying a BRCA pathogenic variant, it is important that genetic testing be considered in advance of surgery, when possible.[68]

In a group of 148 carriers of BRCA1 and BRCA2 pathogenic variants with unilateral breast cancer, 79 of whom underwent CRRM, the risk of CBC was reduced by 91% and was independent of the effect of RRSO. Survival increased among women who underwent CRRM, but this result was likely associated with higher mortality caused by the index cancer or metachronous ovarian cancer in the group that did not have surgery.[69] Data from ten European centers on 550 women (including 202 BRCA carriers) with 3,334 woman-years of follow-up indicated that RRM was highly effective. Bilateral RRMs were carried out on women with a lifetime risk of 25% to 80%, with an average expected incidence rate of 1% per year. No breast cancers occurred in this cohort over the follow-up period, though more than 34 breast cancers would have been expected.[70] A retrospective study of 593 carriers of BRCA1 and BRCA2 pathogenic variants included 105 women with unilateral breast cancer who underwent CRRM and had a 10-year survival rate of 89%, compared with 71% in the group that did not undergo contralateral risk-reducing surgery (P < .001).[4] This study was limited by several factors, such as the lack of information regarding breast cancer screening, grade, and estrogen receptor (ER) status in a large portion of this sample.

A Dutch cohort of 583 patients identified between 1980 and 2011, who had both a BRCA pathogenic variant and a diagnosis of unilateral breast cancer, were evaluated for the effect of CRRM.[71] With a median follow-up of 11.4 years, 242 (42%) of the patients underwent RRM (193 carriers of BRCA1 pathogenic variants and 49 carriers of BRCA2 pathogenic variants) at differing times after their diagnoses. Improved OS was observed in the RRM group when compared with the surveillance group (HR, 0.49; 95% CI, 0.29–0.82), with improvements most pronounced in those diagnosed before age 40 years, with low tumor grade, and non–triple-negative subtype. In an attempt to control for the bias of time to surgery, the authors included a separate evaluation of women who were known to be disease free 2 years after the primary cancer diagnosis (HR, 0.55; 95% CI, 0.32–0.95). Additionally, the group who underwent RRM was more likely to undergo bilateral salpingo-oophorectomy and systemic chemotherapy, which may influence the significance of these survival findings.

A retrospective study of 390 women with early-stage breast cancer who were from families with a known BRCA1/BRCA2 pathogenic variant found a significant improvement in survival for women who underwent RRM when compared with those who chose unilateral mastectomy.[67] Patients were followed for a median of 14.3 years (range, 0.1–20.0 y). A multivariate analysis controlling for age at diagnosis, year of diagnosis, treatment, and other prognostic factors found that CRRM was associated with a 48% reduction in death from breast cancer. This was a relatively small study, and although the authors adjusted for multiple factors, residual confounding factors may have influenced the results.

All of these studies are limited by the biases introduced in relatively small, retrospective studies among very select populations. There is often limited data on potential confounding variables such as socioeconomic status, comorbidities, and access to care. It has been suggested that women who elect to undergo RRM are healthier by virtue of being able to tolerate more extensive surgery. This theory is supported by one study that used Surveillance, Epidemiology, and End Results (SEER) Program data to examine the association between CRRM and outcomes among women with unilateral breast cancer stages I through III. Results showed a reduction in all-cause mortality and breast cancer–specific mortality, and also in noncancer event mortality, a finding that would not be expected to be related to CRRM.[72]

Level of evidence: 3ai

Nipple-sparing mastectomy

The option of nipple-sparing mastectomy (NSM) in carriers of BRCA pathogenic variants undergoing risk-reducing procedures has been controversial because of concerns about increased breast tissue left behind at surgery to keep the NAC viable. The ability to leave behind minimal residual tissue, however, may be related to experience and technique. In a retrospective review of NSM performed in carriers of BRCA pathogenic variants at two hospitals between 2007 and 2014, NSM was performed on 397 breasts in 201 carriers of BRCA pathogenic variants.[73] This study included both unaffected and affected women. Incidental cancers were found in 4 of 150 RRM patients (2.7%) and 2 of 51 cancer patients (3.9%). With a mean follow-up of 32.6 months (range, 1.0–76.0 months), there were four subsequent cancer events that included two patients with axillary recurrences, one patient with a local and distant recurrence 11 months after her original NSM, and one patient who developed a new cancer in the inferior portion of her breast, with no recurrences at the NAC. A study of 177 NSMs performed in 89 carriers of BRCA pathogenic variants between 2005 and 2013 reported similar, excellent local control rates. Sixty-three patients had risk-reducing NSM (median follow-up, 26 months; range, 11–42 months), and 26 patients had NSM and a diagnosis of breast cancer (median follow-up, 28 months; range, 15–43 months). Five patients required further nipple excision. There were no local recurrences or newly diagnosed breast cancers.[74] Although the median follow-up time was short (34 months), NSM appeared to be a safe alternative to traditional mastectomy in appropriately selected patients with BRCA pathogenic variants.[75]

Level of evidence: 3aii

Histopathology of RRM specimens

Studies describing histopathologic findings in RRM specimens from women with BRCA1 or BRCA2 pathogenic variants have been somewhat inconsistent. In two series, proliferative lesions associated with an increased risk of breast cancer (lobular carcinoma in situ, atypical lobular hyperplasia, atypical ductal hyperplasia, and DCIS) were noted in 37% to 46% of women with pathogenic variants who underwent either unilateral or bilateral RRM.[76-78] In these series, 13% to 15% of patients were found to have previously unsuspected DCIS in the prophylactically removed breast. Among 47 cases of RRM or contralateral mastectomies performed in known carriers of BRCA1 or BRCA2 pathogenic variants from Australia, three (6%) cancers were detected at surgery.[79] In general, histopathologic findings in RRM specimens do not impact management.

Utilization

Individual psychological factors play an important role in decision-making about RRM by unaffected women and CRRM in women with unilateral breast cancer. (Refer to the Psychosocial Aspects of Cancer Risk Management for Hereditary Breast and Ovarian Cancer section in the Psychosocial Issues in BRCA1/BRCA2 section of this summary for information about uptake of RRM in BRCA carriers and the Psychosocial Outcome Studies section for information about psychosocial outcomes of RRM.)

Conclusion

In summary:

  • RRM in unaffected women with pathogenic BRCA variants decreases the incidence of breast cancer by approximately 90%, with a less clear impact on breast cancer mortality.
  • RRM in women with unilateral breast cancer (CRRM) has demonstrated decreased incidence of breast cancer and possible improved survival.
  • NSM is an option for RRM, although variation in surgical technique and expertise may leave behind some tissue in the NAC, which may impact the need for subsequent surgery and may increase risk of a subsequent cancer.
  • The predominant histopathologic findings at the time of RRM in unaffected women with pathogenic BRCA variants are proliferative lesions that do not require additional treatment.
Risk-reducing salpingo-oophorectomy (RRSO)

In the general population, removal of both ovaries has been associated with a reduction in breast cancer risk of up to 75%, depending on parity, weight, and age at time of artificial menopause. (Refer to the PDQ summary on Breast Cancer Prevention for more information.) A Mayo Clinic study of 680 women at various levels of familial risk found that in women younger than 60 years who had bilateral oophorectomy, the likelihood of breast cancers developing was reduced for all risk groups.[80] Ovarian ablation, however, is associated with side effects such as hot flashes, impaired sleep habits, vaginal dryness, dyspareunia, and increased risk of osteoporosis and heart disease. A variety of strategies may be necessary to counteract the adverse effects of ovarian ablation.

The evidence for the effect of RRSO on breast cancer has evolved. Early small studies suggested a protective benefit. Initial retrospective studies supported breast cancer and ovarian cancer risk reduction after RRSO in BRCA pathogenic variant–positive women.[81] In support of early small studies,[82,83] a retrospective study of 551 women with disease-associated BRCA1 or BRCA2 variants found a significant reduction in risk of breast cancer (HR, 0.47; 95% CI, 0.29–0.77) and ovarian cancer (HR, 0.04; 95% CI, 0.01–0.16) after RRSO.[81] A prospective, single-institution study of 170 women with BRCA1 or BRCA2 pathogenic variants showed a similar trend. With RRSO, the HR was 0.15 (95% CI, 0.02–1.31) for ovarian, fallopian tube, or primary peritoneal cancer, and 0.32 (95% CI, 0.08–1.2) for breast cancer; the HR for either cancer was 0.25 (95% CI, 0.08–0.74).[84] A prospective, multicenter study of 1,079 women followed up for a median of 30 to 35 months found that while RRSO was associated with reductions in breast cancer risk in both carriers of BRCA1 and BRCA2 pathogenic variants, the risk reduction was more pronounced in BRCA2 carriers (HR, 0.28; 95% CI, 0.08–0.92).[6] A meta-analysis of all reports of RRSO and breast and ovarian/fallopian tube cancers in carriers of BRCA1/BRCA2 pathogenic variants confirmed that RRSO was associated with a significant reduction in breast cancer risk (overall: HR, 0.49; 95% CI, 0.37–0.65; BRCA1: HR, 0.47; 95% CI, 0.35–0.64; BRCA2: HR, 0.47; 95% CI, 0.26–0.84).[85] However, a cohort study of 822 carriers of BRCA1/BRCA2 pathogenic variants conducted in the Netherlands, where carrier screening is performed nationwide, did not observe a reduced risk of breast cancer after RRSO (HR, 1.09; 95% CI, 0.67–1.77).[86] The authors argued that the previous findings were driven by methodological issues including cancer-induced testing bias and immortal person time; the researchers empirically evaluated this by using their own cohort and applying the same assumptions about counting person time from previous studies.[86] In a response, investigators from the U.S. studies analyzed their data using the assumptions of the Dutch study but still observed an inverse association with RRSO and breast cancer risk.[87] In a retrospective cohort of 676 women, carriers having RRSO at the time of breast cancer diagnosis had a reduced risk of breast cancer–specific mortality (HR, 0.38; 95% CI, 0.19–0.77 for BRCA1 carriers and HR, 0.57; 95% CI, 0.23–1.43 for BRCA2 carriers).[88] A subsequent international, multi-institutional study of 3,722 BRCA1 and BRCA2 carriers using a similar methodology showed that oophorectomy performed before age 50 years helped prevent breast cancer in BRCA2 carriers (HR, 0.18; 95% CI, 0.05–0.63; P = .007) but not in BRCA1 carriers.[89] Given these conflicting findings, an additional cohort of 17,917 women who participated in the Prospective Family Study Cohort was assessed, with 1,046 women diagnosed with breast cancer and a median follow-up of 10.7 years; no association between RRSO and breast cancer was observed.[90]

A prospective, multicenter, cohort study of 2,482 carriers of BRCA1/BRCA2 pathogenic variants reported an association of RRSO with a reduction in all-cause mortality (HR, 0.40; 95% CI, 0.26–0.61), breast cancer–specific mortality (HR, 0.44; 95% CI, 0.26–0.76), and ovarian cancer–specific mortality (HR, 0.21; 95% CI, 0.06–0.80).[2] A subsequent meta-analysis confirmed the impact of RRSO on all-cause mortality (HR, 0.32; 95% CI, 0.27–0.38) in carriers of BRCA1 and BRCA2 pathogenic variants, including those with and without a personal history of breast cancer.[91]

Despite discordant findings regarding RRSO and breast cancer risk in the existing literature, aggregate data suggest that there is a benefit, although the magnitude of this benefit may not be fully understood. Further prospective studies are needed to confirm these findings.

Level of evidence: 3ai

Refer to the RRSO section in the Ovarian cancer section of this summary for more information about the effect of RRSO on ovarian cancer risk in carriers of BRCA pathogenic variants.

Chemoprevention
Tamoxifen

Tamoxifen (a synthetic antiestrogen) increases breast-cell growth inhibitory factors and concomitantly reduces breast-cell growth stimulatory factors. The National Surgical Adjuvant Breast and Bowel Project Breast Cancer Prevention Trial (NSABP-P-1), a prospective, randomized, double-blind trial, compared tamoxifen (20 mg/day) with placebo for 5 years in high-risk women (defined by a Gail model risk score >1.66, age >60 y, or lobular carcinoma in situ). Tamoxifen was shown to reduce the risk of invasive breast cancer by 49%. The protective effect was largely confined to ER-positive breast cancer, which was reduced by 69%. The incidence of ER-negative cancer was not significantly reduced.[92] Similar reductions were noted in the risk of preinvasive breast cancer. Reductions in breast cancer risk were noted both among women with a family history of breast cancer and among those without a family history. An increased incidence of endometrial cancers and thrombotic events occurred among women older than 50 years. Interim data from two European tamoxifen prevention trials did not show a reduction in breast cancer risk with tamoxifen after a median follow-up of 48 months [93] or 70 months,[94] respectively. In one trial, however, reduction in breast cancer risk was seen among a subgroup who also used hormone replacement therapy (HRT).[93] These trials varied considerably in study design and populations. (Refer to the PDQ summary on Breast Cancer Prevention for more information.)

Subsequently, the International Breast Cancer Intervention Study 1 (IBIS-1) breast cancer prevention trial randomly assigned 7,154 women between the ages of 35 and 70 years to receive tamoxifen or placebo for 5 years. Eligibility for the trial was based on family history or abnormal benign breast disease. At a median follow-up of 16 years, there was a 29% reduction in risk of breast cancer in the tamoxifen arm (HR, 0.71; 95% CI, 0.60–0.83). There was a 43% reduction in risk for invasive ER-positive breast cancer (HR, 0.66; 95% CI, 0.54–0.81) and a 35% reduction in risk for DCIS (HR, 0.65; 95% CI 0.43–1.00). There was no reduction in risk of invasive ER-negative breast cancer.[95] These findings confirm those of the Breast Cancer Prevention Trial (P-1).[92]

Level of evidence (tamoxifen in a high-risk population): 1aii

A substudy of the NSABP-P-1 trial evaluated the effectiveness of tamoxifen in preventing breast cancer in carriers of BRCA1/BRCA2 pathogenic variants older than 35 years. BRCA2-positive women benefited from tamoxifen to the same extent as BRCA1/BRCA2 pathogenic variant–negative participants; however, tamoxifen use among healthy women with BRCA1 pathogenic variants did not appear to reduce breast cancer incidence. These data must be viewed with caution because of the small number of carriers of pathogenic variants in the sample (8 BRCA1 carriers and 11 BRCA2 carriers).[96]

Level of evidence: 1aii

In contrast to the very limited data on primary prevention in carriers of BRCA1 and BRCA2 pathogenic variants with tamoxifen, several studies have found a protective effect of tamoxifen on the risk of CBC.[97-99] In one study involving approximately 600 carriers of BRCA1/BRCA2 pathogenic variants, tamoxifen use was associated with a 51% reduction in CBC.[97] An update to this report examined 285 carriers of BRCA1/BRCA2 pathogenic variants with bilateral breast cancer and 751 carriers of BRCA1/BRCA2 pathogenic variants with unilateral breast cancer (40% of these patients were included in their initial study). Tamoxifen was associated with a 50% reduction in CBC risk in carriers of BRCA1 pathogenic variants and a 58% reduction in carriers of BRCA2 pathogenic variants. Tamoxifen did not appear to confer benefit in women who had undergone an oophorectomy, although the numbers in this subgroup were quite small.[99] Another study that involved 160 carriers of BRCA1/BRCA2 pathogenic variants demonstrated that tamoxifen use after the treatment of breast cancer with lumpectomy and radiation was associated with a 69% reduction in the risk of CBC.[98] In another study, 2,464 carriers of BRCA1/BRCA2 pathogenic variants with a personal history of breast cancer were identified from three family cohorts. Using both retrospective and prospective data, researchers found a significant decrease in the risk of CBC among women who received adjuvant tamoxifen therapy after their diagnosis. This association persisted after researchers adjusted for age at diagnosis and the ER status of the first cancer. A major limitation of this study is the lack of information on ER status of the first breast cancer in 56% of the women.[100] These studies are limited by their retrospective, case-control designs and the absence of information regarding ER status in the primary tumor.

The STAR trial (NSABP-P-2) included more than 19,000 women and compared 5 years of raloxifene versus tamoxifen in reducing the risk of invasive breast cancer.[101] There was no difference in incidence of invasive breast cancer at a mean follow-up of 3.9 years; however, there were fewer noninvasive cancers in the tamoxifen group. The incidence of thromboembolic events and hysterectomy was significantly lower in the raloxifene group. Detailed quality-of-life data demonstrate slight differences between the two arms.[102] Data regarding efficacy in carriers of BRCA1 or BRCA2 pathogenic variants are not available. (Refer to the PDQ summary on Breast Cancer Prevention for more information about the use of selective ER modulators and aromatase inhibitors in the general population, including postmenopausal women.)

Another case-control study of carriers of pathogenic variants and noncarriers identified through ascertainment of women with bilateral breast cancer found that systemic adjuvant chemotherapy reduced CBC risk among carriers of pathogenic variants (RR, 0.5; 95% CI, 0.2–1.0). Tamoxifen was associated with a nonsignificant risk reduction (RR, 0.7; 95% CI, 0.3–1.8). Similar risk reduction was seen in noncarriers; however, given the higher absolute CBC risk in carriers, there is potentially a greater impact of adjuvant treatment in risk reduction.[103]

The effect of tamoxifen on ovarian cancer risk was studied in 714 carriers of BRCA1 pathogenic variants. All subjects had a prior history of breast cancer; use of tamoxifen was not associated with an increased risk of subsequent ovarian cancer (odds ratio [OR], 0.78; 95% CI, 0.46–1.33).[104]

Reproductive factors

In the general population, breast cancer risk increases with early menarche and late menopause and is reduced at early first full-term pregnancy. (Refer to the PDQ summary on Breast Cancer Prevention for more information.) In the Nurses’ Health Study, the above were risk factors among women who did not have a mother or sister with breast cancer.[105] Among women with a family history of breast cancer, pregnancy at any age appeared to be associated with an increased breast cancer risk, persisting to age 70 years.

One study evaluated risk modifiers among 333 female carriers of a BRCA1 high-risk pathogenic variant. In women with known pathogenic variants of the BRCA1 gene, early age at first live birth and parity of three or more have been associated with a lowered risk of breast cancer. A RR of 0.85 was estimated for each additional birth, up to five or more; however, increasing parity appeared to be associated with an increased risk of ovarian cancer.[106,107] In a case-control study from New Zealand, investigators noted no difference in the impact of parity on the risk of breast cancer between women with a family history of breast cancer and those without a family history of breast cancer.[108]

Studies of the effect of pregnancy on breast cancer risk have revealed complex results. The relationship between parity and breast cancer risk has been inconsistent and may vary between carriers of BRCA1 and BRCA2 pathogenic variants.[109-111] Parity has more consistently been associated with a reduced risk of breast cancer in carriers of BRCA1 pathogenic variants.[109-113] Of note, neither therapeutic nor spontaneous abortions appear to be associated with an increased breast cancer risk.[111,114]

Level of evidence: 4aii

In the general population, breastfeeding has been associated with a slight reduction in breast cancer risk in a few studies, including a large collaborative reanalysis of multiple epidemiologic studies.[115] At least one study suggests that breastfeeding may be protective in carriers of BRCA1 pathogenic variants. In a multicenter, case-control study of 685 carriers of BRCA1 pathogenic variants with breast cancer and 280 carriers of BRCA2 pathogenic variants with breast cancer and 965 carriers without breast cancer drawn from multiple-case families, among carriers of BRCA1 pathogenic variants, breastfeeding for 1 year or more was associated with approximately a 45% reduced risk of breast cancer.[116] No such reduced risk was observed among carriers of BRCA2 pathogenic variants. A second study failed to confirm this association.[114]

Oral contraceptives

There is no consistent evidence that the use of oral contraceptives (OCs) increases the risk of breast cancer in the general population.[117] (Refer to the PDQ summary on Breast Cancer Prevention for more information.)

Although several smaller studies have reported a slightly increased risk of breast cancer with OC use in carriers of BRCA1/BRCA2 pathogenic variants,[118,119] a meta-analysis concluded that the associated risk is not significant with more recent OC formulations.[120] However, OCs formulated before 1975 were associated with an increased risk of breast cancer.[120] A large proportion of patients on whom this meta-analysis was based were drawn from three large studies summarized in Table 12.[121-123]

Table 12. Oral Contraceptive (OC) Use and Breast Cancer Risk in Carriers of BRCA1/BRCA2 Pathogenic Variants
  Kotsopoulos et al. (2014)a [124] Brohet et al. (2007)b [121] Haile et al. (2006)a,c [122] Narod et al. (2002)a [123]
CI = confidence interval.
aReports risk estimates in the form of odds ratios with 95% CIs.
bReports risk estimates in the form of hazard ratios with 95% CIs.
cRisk estimates restricted to carriers of BRCA pathogenic variants younger than 40 years.
Study population BRCA1 carriers with breast cancer N = 2,492 N = 597 N = 195; diagnosis < age 50 y N = 981
BRCA2 carriers with breast cancer Not applicable N = 249 N = 128; diagnosis < age 50 y N = 330
Ever use OC BRCA1 1.18 [CI 1.03–1.36] P = .02 1.47 [CI 1.13–1.91] 0.64 [CI 0.35–1.16] 1.38 [CI 1.11–1.72] P = .003
BRCA2 Not applicable 1.49 [Cl 0.8–2.7] 1.29 [Cl 0.61–2.76] 0.94 [Cl 0.72–1.24]
Age use <20 y BRCA1 1.45 [CI 1.20–1.75] P = .0001 1.41 [Cl 0.99–2.01] 0.84 [Cl 0.45–1.55] 1.36 [Cl 1.11–1.67] P = .003
BRCA2 Not applicable 1.25 [Cl 0.57–2.74] 1.64 [Cl 0.77–3.46] Not reported
Total duration BRCA1 <5 y: 1.14 [CI 0.97–1.35] <9 y: 1.51 [Cl 1.1–2.08] <5 y: 0.61 [Cl 0.31–1.17] <10 y: 1.36 [Cl 1.11–1.67] P = .003
>5 y: 1.22 [CI 1.04–1.49] P = .02
BRCA2 Not applicable <9 y: 2.27 [Cl 1.1–4.65] <5 y: 0.79 [Cl 0.26–2.37] <10 y: 0.82 [Cl 0.56–1.91]
Use before full-term pregnancy BRCA1 Not applicable >4 y: 1.49 [Cl 1.05–2.11] >4 y: 0.69 [Cl 0.41–1.16] Not evaluated
BRCA2 Not applicable >4 y: 2.58 [Cl 1.21–5.49] >4 y: 2.08 [Cl 1.02–4.25] trend per y: 1.11; P trend = .01

When patients are counseled about contraceptive options and preventive actions, the potential impact of OC use on the risk of breast cancer, ovarian cancer, and other health-related effects need to be considered. A number of important issues remain unresolved, including the potential differences between carriers of BRCA1 or BRCA2 pathogenic variants, effect of age and duration of exposure, and effect of OCs on families with highly penetrant early-onset breast cancer.

Level of evidence: 3aii

(Refer to the Oral contraceptives section in the Chemoprevention section of this summary for a discussion of OC use and ovarian cancer in this population.)

Hormone replacement therapy (HRT)

Both observational and randomized clinical trial data suggest an increased risk of breast cancer associated with HRT in the general population.[125-128] The Women’s Health Initiative (WHI) was a randomized controlled trial of approximately 160,000 postmenopausal women that investigated the risks and benefits of dietary interventions and HRT to reduce the incidence of heart disease, breast cancer, colorectal cancer, and fractures. The estrogen-plus-progestin arm of the study, in which more than 16,000 women were randomly assigned to receive combined hormone therapy or placebo, was halted early because health risks exceeded benefits.[127,128] One of the adverse outcomes prompting closure was a significant increase in both total (245 vs. 185 cases) and invasive (199 vs. 150) breast cancers (RR, 1.24; 95% CI, 1.02–1.50; P < .001) in women randomly assigned to receive estrogen and progestin.[128] Results of a follow-up study suggest that the recent reduction in breast cancer incidence, especially among women aged 50 to 69 years, is predominantly related to a decrease in use of combined estrogen plus progestin HRT.[129] HRT-related breast cancers had adverse prognostic characteristics (more advanced stages and larger tumors) when compared with cancers occurring in the placebo group. HRT was also associated with a substantial increase in abnormal mammograms.[128]

Breast cancer risk associated with postmenopausal HRT has been variably reported to be increased [130-132] or unaffected by a family history of breast cancer;[106,133,134] risk did not vary by family history in the meta-analysis.[117] The WHI study has not reported analyses stratified on breast cancer family history, and subjects have not been systematically tested for BRCA1/BRCA2 pathogenic variants.[128] Short-term use of hormones for treatment of menopausal symptoms appears to confer little or no breast cancer risk in the general population.[135]

HRT in carriers of BRCA1/BRCA2 pathogenic variants

The effect of HRT on breast cancer risk among carriers of a BRCA1 or BRCA2 pathogenic variant has been examined in two studies. In a prospective study of 462 carriers of BRCA1 and BRCA2 pathogenic variants, bilateral RRSO (n = 155) was significantly associated with breast cancer risk-reduction overall (HR, 0.40; 95% CI, 0.18–0.92). When carriers of pathogenic variants without bilateral RRSO or HRT were used as the comparison group, HRT use (n = 93) did not significantly alter the reduction in breast cancer risk associated with bilateral RRSO (HR, 0.37; 95% CI, 0.14–0.96).[136] In a matched case-control study of 472 postmenopausal women with BRCA1 pathogenic variants, HRT use was associated with an overall reduction in breast cancer risk (OR, 0.58; 95% CI, 0.35–0.96; P = .03). A nonsignificant reduction in risk was observed both in women who had undergone bilateral oophorectomy and in those who had not. Women taking estrogen alone had an OR of 0.51 (95% CI, 0.27–0.98; P = .04), while the association with estrogen and progesterone was not statistically significant (OR, 0.66; 95% CI, 0.34–1.27; P = .21).[137] A case-control study of 432 matched pairs of postmenopausal women with a BRCA1 pathogenic variant who had a personal history of cancer were compared with unaffected BRCA1 carriers. The use of HRT was not associated with an increased risk of developing breast cancer (OR, 0.80; P = .24).[138] Especially given the differences in estimated risk associated with HRT between observational studies and the WHI, these findings should be confirmed in randomized prospective studies,[139] but they suggest that HRT in carriers of BRCA1/BRCA2 pathogenic variants neither increases breast cancer risk nor negates the protective effect of oophorectomy.

Level of evidence: 3aii

Ovarian cancer

Screening/surveillance

Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Screening for information on screening in the general population and to the PDQ summary Levels of Evidence for Cancer Genetics Studies for information about levels of evidence related to screening and prevention. The latter also outlines the five requirements that must be met before it is considered appropriate to screen for a particular medical condition as part of routine medical practice.

Clinical examination

In the general population, clinical examination of the ovaries has neither the specificity nor the sensitivity to reliably identify early-stage ovarian cancer. No data exist regarding the benefit of clinical examination of the ovaries (bimanual pelvic examination) in women at inherited risk of ovarian cancer.

Level of evidence: None assigned

Transvaginal ultrasound

In the general population, transvaginal ultrasound (TVUS) appears to be superior to transabdominal ultrasound in the preoperative diagnosis of adnexal masses. Both techniques have lower specificity in premenopausal women than in postmenopausal women due to the cyclic menstrual changes in premenopausal ovaries (e.g., transient corpus luteum cysts) that can cause difficulty in interpretation. The randomized prospective Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO-1) found no reduction in mortality with the annual use of combined TVUS and cancer antigen 125 (CA-125) in screening asymptomatic postmenopausal women at general-population risk of ovarian cancer.[140]

Data are limited regarding the potential benefit of TVUS in screening women at inherited risk of ovarian cancer. A number of retrospective studies have reported experience with ovarian cancer screening in high-risk women using TVUS with or without CA-125.[9,141-151] However, there is little uniformity in the definition of high-risk criteria and compliance with screening, and in whether cancers detected were incident or prevalent. One of the largest reported studies included 888 carriers of BRCA1/BRCA2 pathogenic variants who were screened annually with TVUS and CA-125. Ten women developed ovarian cancer; five of the ten developed interval cancers after normal screening results within 3 to 10 months before diagnosis. Five of the ten ovarian cancers were screen-detected incident cases, which had normal screening results within 6 to 14 months before diagnosis. Of these five cases, four were stage IIIB or IV.[141]

A similar study reported the results of annual TVUS and CA-125 in a cohort of 312 high-risk women (152 carriers of BRCA1/BRCA2 pathogenic variants).[143] Of the four cancers that were detected due to abnormal TVUS and CA-125, all four patients were symptomatic, and three had advanced-stage disease. Annual screening of carriers of BRCA1/BRCA2 pathogenic variants with pelvic ultrasound, TVUS, and CA-125 failed to detect early-stage ovarian cancer among 241 carriers of BRCA1/BRCA2 pathogenic variants in a study from the Netherlands.[152] Three cancers were detected over the course of the study, all advanced stage IIIC disease.[152] Finally, a study of 1,100 moderate- and high-risk women who underwent annual TVUS and CA-125 reported that 10 of 13 ovarian tumors were detected due to screening. Only 5 of 10 were stage I or II.[142] There are limited data related to the efficacy of semiannual screening with TVUS and CA-125.[9,150]

In the United Kingdom Familial Ovarian Cancer Screening Study, 3,563 women with an estimated 10% or higher lifetime risk of ovarian cancer were screened with annual ultrasound and serum CA-125 measurements for a mean of 3.2 years. Four of 13 screen-detected cancers were stage I or II. Women screened within the previous year were less likely to have higher than stage IIIC disease; there was also a trend towards better rates of optimal cytoreduction and improved OS. Furthermore, most of the cancers occurred in women with known ovarian cancer susceptibility genes, identifying a cohort at highest cancer risk for consideration of screening.[153] Phase II of this study increased the frequency of screening to every 4 months; the impact of this is not yet available.

The first prospective study of TVUS and CA-125 with survival as the primary outcome was completed in 2009. Of the 3,532 high-risk women screened, 981 were carriers of BRCA pathogenic variants, 49 of whom developed ovarian cancer. The 5- and 10-year survival rates were 58.6% (95% CI, 50.9%–66.3%) and 36% (95% CI, 27–45), respectively, and there were no differences in survival rates between carriers and noncarriers. A major limitation of the study was the absence of a control group. Despite limitations, this study suggests that annual surveillance by TVUS and CA-125 level appears to be ineffective in detecting tumors at an early stage to substantially influence survival.[154]

Level of evidence: 4

Serum CA-125

Serum CA-125 screening for ovarian cancer in high-risk women has been evaluated in combination with TVUS in a number of retrospective studies, as described in the previous section.[9,141-150]

The National Institutes of Health (NIH) Consensus Statement on Ovarian Cancer recommended against routine screening of the general population for ovarian cancer with serum CA-125. (Refer to the Prostate, Lung, Colorectal and Ovarian [PLCO] Cancer Screening Trial: Single-threshold CA-125 levels and TVU section in the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Screening for more information.) The NIH Consensus Statement did, however, recommend that women at inherited risk of ovarian cancer undergo TVUS and serum CA-125 screening every 6 to 12 months, beginning at age 35 years.[155] The Cancer Genetics Studies Consortium task force has recommended that female carriers of a BRCA1 pathogenic variant undergo annual or semiannual screening using TVUS and serum CA-125 levels, beginning at age 25 to 35 years.[11] Both recommendations are based solely on expert opinion and best clinical judgment.

Level of evidence: 5

Other candidate ovarian cancer biomarkers

The need for effective ovarian cancer screening is particularly important for women carrying BRCA1 and BRCA2 pathogenic variants, and the mismatch repair (MMR) genes (e.g., MLH1, MSH2, MSH6, PMS2), disorders in which the risk of ovarian cancer is high. There is a special sense of urgency for carriers of BRCA1 pathogenic variants, in whom cumulative lifetime risks of ovarian cancer may exceed 40%.

Thus, it is expected that many new ovarian cancer biomarkers (either singly or in combination) will be proposed as ovarian cancer screening strategies during the next 5 to 10 years. While this is an active area of research with a number of promising new biomarkers in early development, at present, none of these biomarkers alone or in combination have been sufficiently well studied to justify their routine clinical use for screening purposes, either in the general population or in women at increased genetic risk.

Before information related to emerging ovarian cancer biomarkers is addressed, it is important to consider the several steps that are required to develop and, more importantly, validate a new biomarker. One useful framework was published by the National Cancer Institute Early Detection Research Network investigators.[156] They indicated that the goal of a cancer-screening program is to detect tumors at an early stage so that treatment is likely to be successful. The gold standard by which such programs are judged is whether the death rate from the cancer for which screening is performed is reduced among those being screened. In addition, the screening test must be sufficiently noninvasive and inexpensive to allow widespread use in the screening population. Maintaining high test specificity (i.e., few false-positive results) is essential for a population screening test, because even a low false-positive rate results in many people having to undergo unnecessary, costly diagnostic procedures and psychological stress. It is likely that the use of several cancer biomarkers will be required for a screening test to be both sensitive and specific.

Furthermore, a clinically useful test must have a high PPV (a parameter derived from sensitivity, specificity, and disease prevalence in the screened population). Practically speaking, a biomarker with a PPV of 10% implies that ten surgical procedures would be required to identify one case of ovarian cancer; the remaining nine surgeries would represent false-positive test findings. In general, the ovarian cancer research community considers biomarkers with a PPV of less than 10% to be clinically unacceptable, given the morbidity related to bilateral salpingo-oophorectomy. Finally, while novel biomarkers may be present in the sera of women with advanced ovarian cancer (who represent most cases analyzed in the early phases of biomarker development), they may or may not be detectable in women with early-stage disease, which is essential if the screening test is to be clinically useful.

It has been suggested that there are five general phases in biomarker development and validation:

Phase 1 — Preclinical exploratory studies

  • Identify potentially discriminating biomarkers.
  • Usually done by comparing gene over- or under-expression in the tumor when compared with normal tissue.
  • Because many exploratory analyses in large numbers of genes are performed at this stage, one or more may seem to have good discriminating ability between cancers and normal tissue by random chance alone.

Phase 2 — Clinical assay development for clinical disease

  • Develop a clinical assay that can be obtained on noninvasively obtained samples (e.g., a blood specimen).
  • Often the test targets the protein product of one of the genes found to be of interest in phase 1.
  • The goal is to describe the performance characteristics of the assay for distinguishing between subjects with and without cancer. At this point, the assay should be in its final configuration and remain stable throughout the following phases.
  • IMPORTANT: Because the case subjects in a phase 2 study already have cancer, with assay results obtained at the time of disease diagnosis, one cannot determine whether disease can be detected early with a given biomarker.

Phase 3 — Retrospective longitudinal repository studies

  • Compare clinical specimens collected from cancer case subjects before their clinical diagnosis with specimens from subjects who have not developed cancer.
  • Evaluate, as a function of time before clinical diagnosis, the biomarker’s ability to detect preclinical disease.
  • Define the criteria for a positive screening test in preparation for phase 4.
  • Explore the influence of other patient characteristics (e.g., age, gender, smoking status, medication use) on the ability of the biomarker to discriminate between those with and without preclinical disease.

Phase 4 — Prospective screening studies

  • Determine the operating characteristics of the biomarker-based screening test in a population for which the test is intended.
  • Measure the detection rate (number of abnormal tests among all those with the disease) and the false-positive rate (the number of abnormal tests among all those who do not have the disease).
  • Evaluate whether the cancers detected by the test are being found at an early stage, a point at which treatment is more likely to be curative.
  • Assess whether the test is acceptable in a population of individuals for whom it is intended. Will subjects comply with the test schedule and results?

Phase 5 — Cancer control studies

  • Ideally, conduct randomized controlled clinical trials in clinically relevant populations, in which one arm is subjected to screening and appropriate intervention if screen-positive, while the other arm is not screened.
  • Determine whether the death rate of the cancer being screened for is reduced among those who use the screening test.
  • Obtain information about the costs of screening and treatment of screen-detected cancers.

Finally, for a validated biomarker test to be considered appropriate for use in a particular population, it must have been evaluated in that specific population without prior selection of known positives and negatives. In addition, the test must demonstrate clinical utility, that is, a positive net balance of benefits and risks associated with the application of the test. These may include improved health outcomes and net psychosocial and economic benefits.[157]

Ovarian cancer poses a unique challenge relative to the potential impact of false-positive test results. There are no reliable noninvasive diagnostic tests for early-stage disease, and clinically significant early-stage cancer may not be grossly visible at the time of exploratory surgery.[158] Consequently, it is likely that some patients will be reassured that their abnormal test does not indicate the presence of cancer only by having their ovaries and fallopian tubes surgically removed and examined microscopically. High test specificity (i.e., a very low false-positive rate) is required to avoid unnecessary surgery and induction of premature menopause women with in false-positive results.

Variations on CA-125
CA-125 plus an ovarian cancer symptom index

An ovarian cancer symptom index for predicting the presence of cancer was evaluated in 75 cases and 254 high-risk controls (carriers of BRCA pathogenic variants or women with a strong family history of breast and ovarian cancer).[159] Women had a positive symptom index if they reported any of the predefined symptoms (bloating or increase in abdominal size, abdominal or pelvic pain, and difficulty eating or feeling full quickly) more than 12 times per month, occurring only within the prior 12 months. CA-125 values greater than 30 U/mL were considered abnormal. The symptom index independently predicted the presence of ovarian cancer, after controlling for CA-125 levels (P < .05). The combination of an elevated CA-125 and a positive-symptom index correctly identified 89.3% of the cases. The symptom index correlated with the presence of cancer in 50% of the affected women who did not have elevated CA-125 levels, but 11.8% of the high-risk controls without cancer also had a positive-symptom index. The authors suggested that a composite index that included both CA-125 and the symptom index had better performance characteristics than either test used alone, and that this strategy might be used as a first screen in a multistep screening program. Additional test performance validation and determination of clinical utility are required in unselected screening populations.

Level of evidence: 5

Risk of ovarian cancer algorithm

A novel modification of CA-125 screening is based on the hypothesis that rising CA-125 levels over time may provide better ovarian cancer screening performance characteristics than simply classifying CA-125 as normal or abnormal based on an arbitrary cut-off value. This has been implemented in the form of the risk of ovarian cancer algorithm (ROCA), an investigational statistical model that incorporates serial CA-125 test results and other covariates into a computation that produces an estimate of the likelihood that ovarian cancer is present in the screened subject. The first report of this strategy, based on reanalysis of 5,550 average-risk women from the Stockholm Ovarian Cancer screening trial, suggested that ovarian cancer cases and controls could be distinguished with 99.7% sensitivity, 83% specificity, and a PPV of 16%. That PPV represents an eightfold increase over the 2% PPV reported with a single measure of CA-125.[160] This report was followed by applying the ROCA to 33,621 serial CA-125 values obtained from the 9,233 average-risk postmenopausal women in a prospective British ovarian cancer screening trial.[161] The area under the receiver operator curve increased from 84% to 93% (P = .01) for ROCA compared with a fixed CA-125 cutoff. These observations represented the first evidence that preclinical detection of ovarian cancer might be improved using this screening strategy. A prospective study of 13,000 normal volunteers aged 50 years and older in England used serial CA-125 values and the ROCA to stratify participants into low, intermediate, and elevated risk subgroups.[162] Each had its own prescribed management strategy, including TVUS and repeat CA-125 either annually (low risk) or at 3 months (intermediate risk). Using this protocol, ROCA was found to have a specificity of 99.8% and a PPV of 19%.

Two prospective trials in England utilized the ROCA. The United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) randomly assigned normal-risk women to either (1) no screening, (2) annual ultrasound, or (3) multimodal screening (N = 202,638; accrual completed; follow-up ends in 2014), and the U.K. Familial Ovarian Cancer Screening Study (UKFOCSS) targeted high-risk women (accrual completed). There are also two high-risk cohorts using the ROCA under evaluation in the United States: the Cancer Genetics Network ROCA Study (N = 2,500; follow-up complete; analysis underway) and the Gynecologic Oncology Group Protocol 199 (GOG-0199; enrollment complete; follow-up ended in 2011).[163] Thus, additional data regarding the utility of this currently investigational screening strategy will become available.

Level of evidence: 4

Miscellaneous new markers

A wide array of new candidate ovarian cancer biomarkers has been described during the past decade, e.g., HE4; mesothelin; kallikreins 6, 10, and 11; osteopontin; prostasin; M-CSF; OVX1; lysophosphatidic acid; vascular endothelial growth factor B7-H4; and interleukins 6 and 8.[164-166] These have been singly studied, in combination with CA-125, or in various other permutations. Most of the study populations are relatively small and comprise highly selected, known ovarian cancer cases and healthy controls of the type evaluated in early biomarker development phases 1 and 2. Results have not been consistently replicated in multiple studies; presently, none are considered ready for widespread clinical application.

Level of evidence: 5

Proteomics

Initially, mass spectroscopy of serum proteins was combined with complex analytic algorithms to identify protein patterns that might distinguish between ovarian cancer cases and controls.[167] This approach assumed that pattern recognition alone would be sufficient to permit such discrimination, and that identification of the specific proteins responsible for the patterns identified was not required. This strategy was modified, using similar laboratory tools, to identify finite numbers of specific known serum markers that may be used in place of, or in conjunction with, CA-125 measurements for the early detection of cancer.[168] These studies [166,169] have generally been small case-control studies that are limited by sample size and the number of early-stage cancer cases included. Further evaluation is needed to determine whether any additional markers identified in this fashion have clinical utility for the early detection of ovarian cancer in the unselected clinical population of interest.

Level of evidence: 5

Multiplex biomarker assays for ovarian cancer screening

Because individual biomarkers have not met the criteria for an effective screening test, it has been suggested that it may be necessary to combine multiple ovarian cancer biomarkers to obtain satisfactory screening test results. This strategy was employed to quantitatively analyze six serum biomarkers (leptin, prolactin, osteopontin, insulin-like growth factor II, macrophage inhibitory factor, and CA-125), using a multiplex, bead-based platform.[170] A similar assay was available commercially under the trade name OvaSure until its voluntary withdrawal from the market by the manufacturer.[Response to FDA Warning Letter]

The cases in this study were newly diagnosed ovarian cancer patients who had blood collected just before surgery: 36 were stage I and II; 120 were stage III and IV. The controls were healthy age-matched individuals who had not developed ovarian cancer within 6 months of blood draw. Neither cases nor controls in this study were well characterized regarding their familial and/or genetic risk status, but they have been suggested to comprise a high-risk population. First, 181 controls and 113 ovarian cancer cases were tested to determine the initial panel of biomarkers that best discriminated between cases and controls (training set). The resulting panel was applied to an additional 181 controls and 43 ovarian cancer cases (test set). Pooling both early- and late-stage ovarian cancer across the combined training and test sets, performance characteristics were reported as a sensitivity of 95.3% and a specificity of 99.4%, with a PPV of 99.3% and a negative predictive value of 99.2%, using a formula that assumed an ovarian cancer prevalence of about 50%, as seen in the highly selected research population.

To avoid biases that may make test performance appear to be better than it really is, combining training populations and test populations in analyses of this sort is generally not recommended.[171] The most appropriate prevalence to use is the disease prevalence in the unselected population to be screened. The prevalence of ovarian cancer in the general population is 1 in 2,500. In a correction to their manuscript,[170] the authors assumed that the prevalence of ovarian cancer in the screened population was 1 in 2,500 (0.04%) and recalculated the PPV to be only 6.5%. On that basis, the investigators have retracted their claim that this test is suitable for population screening. If this test were used in patients at increased risk of ovarian cancer, the actual prevalence in such a target population is likely to be higher than that observed in the general population, but well below the assumed 50% figure used in the published analysis. This revised PPV of 6.5% indicates that approximately 1 in 15 women with a positive test would in fact have ovarian cancer, and only a fraction of those with ovarian cancer would be stages I or II. The remaining 14 positive tests would represent false-positives, and these women would be at risk of exposure to needless anxiety and potentially morbid diagnostic procedures, including bilateral salpingo-oophorectomy.

Viewed in the context of the criteria previously described,[156] this assay would be classified as phase 2 in its development. While this appears to be a promising avenue of ovarian cancer screening research, additional validation is required, particularly in an unselected population representative of the clinical screening population of interest. A position statement by the Society of Gynecologic Oncologists regarding this assay indicated, “It is our opinion that additional research is needed to validate the test’s effectiveness before offering it to women outside of the context of a research study conducted with appropriate informed consent under the auspices of an institutional review board.”

Level of evidence: 5

Risk-reducing surgery
RRSO

Numerous studies have found that women with an inherited risk of breast and ovarian cancer have a decreased risk of ovarian cancer after RRSO. (Refer to the RRSO section in the Breast cancer section of this summary for more information.) A retrospective study of 551 women with BRCA1 or BRCA2 pathogenic variants found a significant reduction in risk of breast cancer (HR, 0.47; 95% CI, 0.29–0.77) and ovarian cancer (HR, 0.04; 95% CI, 0.01–0.16) after bilateral oophorectomy.[81] A prospective, single-institution study of 170 women with BRCA1 or BRCA2 pathogenic variants showed a similar trend.[84] With oophorectomy, the HR was 0.15 (95% CI, 0.02–1.31) for ovarian, fallopian tube, or primary peritoneal cancer, and 0.32 (95% CI, 0.08–1.2) for breast cancer; the HR for either cancer was 0.25 (95% CI, 0.08–0.74). A prospective multicenter study of 1,079 women who were followed up for a median of 30 to 35 months found that RRSO is highly effective in reducing ovarian cancer risk in carriers of BRCA1 and BRCA2 pathogenic variants. This study also showed that RRSO was associated with reductions in breast cancer risk in both carriers of BRCA1 and BRCA2 pathogenic variants; however, the breast cancer risk reduction was more pronounced in BRCA2 carriers (HR, 0.28; 95% CI, 0.08–0.92).[6] In a case-control study in Israel, bilateral oophorectomy was associated with reduced ovarian/peritoneal cancer risks (OR, 0.12; 95% CI, 0.06–0.24).[172] A meta-analysis of all reports of RRSO and breast and ovarian/fallopian tube cancer in carriers of BRCA1/BRCA2 pathogenic variants confirmed that RRSO was associated with a significant reduction in risk of ovarian or fallopian tube cancer (HR, 0.21; 95% CI, 0.12–0.39). The study also found a significant reduction in risk of breast cancer (overall: HR, 0.49; 95% CI, 0.37–0.65; BRCA1: HR, 0.47; 95% CI, 0.35–0.64; BRCA2: HR, 0.47; 95% CI, 0.26–0.84).[85] Subsequently, a matched case-control study of 2,854 pairs of women with a BRCA1 or BRCA2 pathogenic variant with or without breast cancer showed a greater breast cancer risk reduction with surgical menopause (OR, 0.52; 95% CI, 0.40–0.66) than with natural menopause (OR, 0.81; 95% CI, 0.62–1.07). This study also reported a highly significant reduction in breast cancer risk among women who had an oophorectomy after natural menopause (OR, 0.13; 95% CI, 0.02–0.54; P = .006).[173] Another study of 5,783 women with BRCA1 or BRCA2 pathogenic variants who were followed up for an average of 5.6 years reported that 68 of 186 women who developed either ovarian, fallopian, or peritoneal cancer had died. The HR for these cancers with bilateral oophorectomy was 0.20 (95% CI, 0.13–0.30; P = .001). In carriers of BRCA pathogenic variants without a history of cancer, the HR for all-cause mortality to age 70 years associated with oophorectomy was 0.23 (95% CI, 0.13–0.39; P < .001).[7] Among studies with 50 or more subjects, prevalence ranged from 2.3% to 11%. Some of the variation in prevalence is likely due to differences in surgical technique, pathologic handling of the tissues, and age at RRSO. In the GOG 199 study of 966 high-risk women, the incidence of occult cancer was highest among carriers of BRCA1 pathogenic variants (4.6%), followed by carriers of BRCA2 pathogenic variants (3.5%), versus only 0.5% of noncarriers. The odds of an occult pathologic finding was fourfold higher among postmenopausal women.[174]

In addition to a reduction in risk of ovarian and breast cancer, RRSO may also significantly improve OS and breast and ovarian cancer–specific survival. A prospective cohort study of 666 women with germline pathogenic variants in BRCA1 and BRCA2 found an HR for overall mortality of 0.24 (95% CI, 0.08–0.71) in women who had RRSO compared with women who did not.[175] This study provides the first evidence to suggest a survival advantage among women undergoing RRSO.

Studies on the degree of risk reduction afforded by RRSO have begun to clarify the spectrum of occult cancers discovered at the time of surgery. Primary fallopian tube cancers, primary peritoneal cancers, and occult ovarian cancers have all been reported. Several case series have reported a prevalence of malignant findings among carriers of pathogenic variants undergoing risk-reducing oophorectomy. Among studies with 50 or more subjects, prevalence ranged from 2.3% to 11%.[9,84,176-182] Some of the variation in prevalence probably results from differences in surgical technique, pathologic handling of the tissues, and age at RRSO. In the GOG 199 study of 966 high-risk women, the incidence of occult cancer was highest in carriers of BRCA1 pathogenic variants (4.6%), followed by carriers of BRCA2 pathogenic variants (3.5%), versus only 0.5% of noncarriers. The odds of an occult pathologic finding was fourfold higher among postmenopausal women.[174]

In addition to occult cancers, premalignant lesions have also been described in fallopian tube tissue removed for prophylaxis. In one series of 12 women with BRCA1 pathogenic variants undergoing risk-reducing surgery, 11 had hyperplastic or dysplastic lesions identified in the tubal epithelium. In several of the cases the lesions were multifocal.[183] These pathologic findings are consistent with the identification of germline BRCA1 and BRCA2 pathogenic variants in women affected with both tubal and primary peritoneal cancers.[180,184-189] One study suggests a causal relationship between early tubal carcinoma, or tubal intraepithelial carcinoma, and subsequent invasive serous carcinoma of the fallopian tube, ovary, or peritoneum.[190] (Refer to the Pathology of ovarian cancer section of this summary for more information.)

These findings support the inclusion of fallopian tube cancers, which account for less than 1% of all gynecologic cancers in the general population, as a component of hereditary ovarian cancer syndrome and necessitate removal of the fallopian tubes at the time of risk-reducing surgery. There is clear evidence that RRSO must include routine collection of peritoneal washings and careful adherence to comprehensive pathologic evaluation of the entire adnexa with the use of serial sectioning.[182,191,192]

The peritoneum, however, appears to remain at low risk for the development of a Müllerian-type adenocarcinoma, even after oophorectomy.[193-197] Of the 324 women from the Gilda Radner Familial Ovarian Cancer Registry who underwent risk-reducing oophorectomy, 6 (1.8%) subsequently developed primary peritoneal carcinoma. No period of follow-up was specified.[198] Among 238 individuals in the Creighton Registry with BRCA1/BRCA2 pathogenic variants who underwent risk-reducing oophorectomy, 5 subsequently developed intra-abdominal carcinomatosis (2.1%). Of note, all five of these women had BRCA1 pathogenic variants.[199] A study of 1,828 women with a BRCA1 or BRCA2 pathogenic variant found a 4.3% risk of primary peritoneal cancer at 20 years after RRSO.[200]

Data are limited regarding outcomes of carriers of BRCA1 and BRCA2 pathogenic variants who are found to have occult lesions at the time of RRSO. In a multi-institution study of 32 women with either invasive carcinoma (n = 15) or serous tubal intraepithelial carcinoma (STIC) (n = 17), 47% of women with invasive cancer had a recurrence at a median time of 32.5 months, with an OS rate of 73%.[201] For women with intraepithelial lesions, one patient (approximately 6%) had a recurrence at 43 months, suggesting a different disease process between the two entities. Another study confirmed the malignant potential of STIC lesions. While 3 of 243 women (1.2%) with benign pathology at RRSO subsequently developed primary peritoneal carcinoma, 2 of 9 women (22%) with STIC developed high-grade pelvic serous carcinoma after a median follow-up time of 63 months.[202]

Given current ovarian cancer screening limitations and the high risk of disease associated with BRCA1 and BRCA2 pathogenic variants, NCCN guidelines recommend RRSO as an effective risk-reducing option; RRSO is recommended between the ages of 35 and 40 years and upon completion of childbearing.[33] Optimal timing of RRSO is individualized, but evaluating a woman's risk of ovarian cancer based on pathogenic variant status can be helpful in the decision-making process. In a large study of U.S. BRCA1 and BRCA2 families, age-specific cumulative risk of ovarian cancer at age 40 years was 4.7% for carriers of BRCA1 pathogenic variants and 1.9% for carriers of BRCA2 pathogenic variants.[203] In a combined analysis of 22 studies of BRCA1 and BRCA2 pathogenic variant carriers, risk of ovarian cancer for BRCA1 pathogenic variant carriers increased the most from age 40 years to age 50 years; the risk of ovarian cancer for BRCA2 pathogenic variant carriers increased sharply from age 50 years to age 60 years.[204] In a population-based study of BRCA pathogenic variants in ovarian cancer patients, patients with BRCA2 variants had a significantly later age of onset of ovarian cancer than patients with BRCA1 variants (57.3 years [range, 40–72] vs. 52.6 years [range, 31–78]).[205] A Markov model studying a hypothetical cohort of patients with BRCA1/BRCA2 pathogenic variants found that the optimal strategy for improving survival was to proceed with RRSO as early as possible.[206] In this model, the ovarian cancer cluster region may be associated with lower life expectancy estimates when compared with the breast cancer cluster regions or the non-cluster regions; however, this did not significantly change life expectancy outcomes unless RRSO was significantly delayed. In summary, women with BRCA1 pathogenic variants may consider RRSO for ovarian cancer risk reduction at an earlier age than women with BRCA2 pathogenic variants; however, women with BRCA2 variants may still consider early RRSO for breast cancer risk reduction.

The role of concomitant hysterectomy at the time of RRSO in carriers of BRCA1/BRCA2 pathogenic variants is controversial. There is concern that a small portion of the proximal fallopian tube remains when hysterectomy is not performed, thereby resulting in a residual increased risk of fallopian tube cancer. However, several studies that have examined fallopian tube cancers indicate that the vast majority of these cancers occur in the distal or midportion of the fallopian tube, suggesting that the occurrence of proximal fallopian tube cancer would be a very unlikely event. Some reports have suggested an increased incidence of uterine carcinoma in carriers of pathogenic variants,[207] whereas others have not confirmed an elevated risk of serous uterine cancer.[208] A prospective study of 857 women suggested that any increased incidence of uterine cancer appeared to be among carriers of BRCA1 pathogenic variants who used tamoxifen;[209] this was confirmed by the same group in a later study of 4,456 carriers of BRCA1/BRCA2 pathogenic variants.[210] Even with tamoxifen use, the excess risk of endometrial cancer was small, with a 10-year cumulative risk of 2%.[210] In addition, the use of tamoxifen can now be minimized, given the options of raloxifene (which does not increase the risk of uterine cancer) and aromatase inhibitors for breast cancer prevention in postmenopausal women. Therefore, on the basis of the current understanding of the risk of uterine cancer in carriers of BRCA pathogenic variants, there is not a singularly compelling reason to consider hysterectomy at the time of RRSO to reduce the risk of uterine cancer. Concomitant hysterectomy does offer the advantage of simplifying the hormone replacement regimen for carriers of BRCA pathogenic variants who choose to take hormones. After hysterectomy, women can take estrogen alone (which does not increase the risk of breast cancer), without progestins, thereby eliminating the risk of postmenopausal bleeding.

Studies indicate that removal of the uterus is not necessary as a risk-reducing procedure. No increased BRCA pathogenic variant prevalence was seen among 200 Jewish women with endometrial carcinoma or 56 unselected women with uterine papillary serous carcinoma.[208,211] However, small studies have reported that uterine papillary serous carcinoma may be part of the BRCA-associated spectrum of disease.[207,212,213] The cumulative risk of endometrial cancer among carriers of BRCA pathogenic variants with ER-positive breast cancer treated with tamoxifen may be an additional factor to consider when counseling this population about risk-reducing hysterectomy.[209,214] Hysterectomy might also be considered in young, unaffected carriers of BRCA pathogenic variants who may want to use HRT but for whom hysterectomy would offer a simplified regimen of estrogen alone. In counseling a carrier of a BRCA pathogenic variant about optimal risk-reducing surgical options, aggregate data suggest that the risk from residual tubal tissue after RRSO is the least compelling reason to suggest hysterectomy. Therefore, in the absence of tamoxifen use or other underlying uterine or cervical problems, hysterectomy is not a routine component of RRSO for BRCA carriers.

For women who are premenopausal at the time of surgery, the symptoms of surgical menopause (e.g., hot flashes, mood swings, weight gain, and genitourinary complaints) can cause a significant impairment in their quality of life. To reduce the impact of these symptoms, providers have often prescribed a time-limited course of systemic HRT after surgery. (Refer to the Hormone replacement therapy in carriers of BRCA1/BRCA2 pathogenic variants section of this summary for more information.)

Studies have examined the effect of RRSO on quality of life (QOL). One study examined 846 high-risk women of whom 44% underwent RRSO and 56% had periodic screening.[215] Of the 368 carriers of BRCA1/BRCA2 pathogenic variants, 72% underwent RRSO. No significant differences were observed in QOL scores (as assessed by the Short Form-36) between those with RRSO or screening or compared with the general population; however, women with RRSO had fewer breast and ovarian cancer worries (P < .001) and more favorable cancer risk perception (P < .05) but more endocrine symptoms (P < .001) and worse sexual functioning (P < .05). Of note, 37% of women used HRT after RRSO, although 62% were either perimenopausal or postmenopausal.[215] Researchers then examined 450 premenopausal high-risk women who had chosen either RRSO (36%) or screening (64%). Of those in the RRSO group, 47% used HRT. HRT users (n = 77) had fewer vasomotor symptoms than did nonusers (n = 87; P < .05), but they had more vasomotor symptoms than did women in the screening group (n = 286). Likewise, women who underwent RRSO and used HRT had more sexual discomfort due to vaginal dryness and dyspareunia than did those in the screening group (P < .01). Therefore, while such symptoms are improved via HRT use, HRT is not completely effective, and additional research is warranted to address these important issues.

The long-term nononcologic effects of RRSO in carriers of BRCA1/BRCA2 pathogenic variants are unknown. In the general population, RRSO has been associated with increased cardiovascular disease, dementia, death from lung cancer, and overall mortality.[216-220] When age at oophorectomy has been analyzed, the most detrimental effect has been seen in women who undergo RRSO before age 45 years and do not take estrogen replacement therapy.[216] Carriers of BRCA1/BRCA2 pathogenic variants undergoing RRSO may have an increased risk of metabolic syndrome.[221] RRSO has also been associated with an improvement in short-term mortality in this population.[175] The benefits related to cancer risk reduction after RRSO are clear, but further data on the long-term nononcologic risks and benefits are needed.

Bilateral salpingectomy

Bilateral salpingectomy has been suggested as an interim procedure to reduce risk in carriers of BRCA pathogenic variants.[222,223] There are no data available on the efficacy of salpingectomy as a risk-reducing procedure. The procedure preserves ovarian function and spares the premenopausal patient the adverse effects of a premature menopause. The procedure can be performed using a minimally invasive approach, and a subsequent bilateral oophorectomy could be deferred until the patient approaches menopause. While the data make a compelling argument that some pelvic serous cancers in carriers of BRCA pathogenic variants originate in the fallopian tube, some cancers clearly arise in the ovary. Furthermore, bilateral salpingectomy could give patients a false sense of security that they have eliminated their cancer risk as completely as if they had undergone a bilateral salpingo-oophorectomy. A small study of 14 young carriers of BRCA pathogenic variants documented the procedure as feasible.[224] However, efficacy and impact on ovarian function was not assessed in this study. Future prospective trials are needed to establish the validity of the procedure as a risk-reducing intervention.

In a statistical Markov model using Monte Carlo simulation, risk-reducing salpingectomy with delayed oophorectomy was a cost-effective strategy considering quality-adjusted life expectancy for women with pathogenic variants in BRCA1/BRCA2.[225] Another study modeling ovarian cancer risk and effects of RRSO and salpingectomy found that the difference in estimated ovarian cancer risk is small when salpingectomy is performed on women of childbearing age and oophorectomy is performed 5 to 10 years later.[226]

Prospective studies are under way evaluating the impact of bilateral salpingectomy with delayed oophorectomy on patient satisfaction and the reduction in ovarian cancer.[227]

Chemoprevention
Oral contraceptives

OCs have been shown to have a protective effect against ovarian cancer in the general population.[228] Several studies, including a large, multicenter, case-control study, showed a protective effect,[125,229-232] while one population-based study from Israel failed to demonstrate a protective effect.[233]

There has been great interest in determining whether a similar benefit extends to women who are at increased genetic risk of ovarian cancer. A multicenter study of 799 ovarian cancer patients with BRCA1 or BRCA2 pathogenic variants, and 2,424 control patients without ovarian cancer but with a BRCA1 or BRCA2 pathogenic variant, showed a significant reduction in ovarian cancer risk with use of OCs (OR, 0.56; 95% CI, 0.45–0.71). Compared with never-use of OCs, duration up to 1 year was associated with an OR of 0.67 (95% CI, 0.50–0.89). The OR for each year of OC use was 0.95 (95% CI, 0.92–0.97), with a maximum observed protection at 3 years to 5 years of use.[232] This study included women from a prior study by the same authors and confirmed the results of that prior study.[125] A population-based case-control study of ovarian cancer did not find a protective benefit of OC use in carriers of BRCA1 or BRCA2 pathogenic variants (OR, 1.07 for ≥5 years of use), although they were protective, as expected, among noncarriers (OR, 0.53 for ≥5 years of use).[233] A small, population-based, case-control study of 36 carriers of BRCA1 pathogenic variants, however, observed a similar protective effect in both carriers of pathogenic variants and noncarriers (OR, approximately 0.5).[231] A larger case-control study of women with pathogenic variants in BRCA1 demonstrated maximum benefit after 5 years of OC use, while women with pathogenic variants in BRCA2 seemed to reach maximum benefit after 3 years of OC use.[234] A multicenter study of subjects drawn from numerous registries observed a protective effect of OCs among the 147 carriers of BRCA1 or BRCA2 pathogenic variants, with ovarian cancer compared with the 304 matched carriers of pathogenic variants without cancer (OR, 0.62 for ≥6 years of use).[230] Finally, a meta-analysis of 18 studies that included 13,627 carriers of BRCA pathogenic variants, 2,855 of whom had breast cancer and 1,503 of whom had ovarian cancer, reported a significantly reduced risk of ovarian cancer (summary RR, 0.50; 95% CI, 0.33–0.75) associated with OC use. The authors also reported significantly higher risk reductions with longer duration of OC use (36% reduction in risk for each additional 10 years of OC use). There was no association with breast cancer risk and use of OC pills formulated after 1975.[120]

Level of evidence: 3aii

(Refer to the Oral contraceptives section in the Reproductive factors section of this summary for a discussion of OC use and breast cancer in this population.)

Reproductive factors

It has been suggested that incessant ovulation, with repetitive trauma and repair to the ovarian epithelium, increases the risk of ovarian cancer. In epidemiologic studies in the general population, physiologic states that prevent ovulation have been associated with decreased risk of ovarian cancer. It has also been suggested that chronic overstimulation of the ovaries by luteinizing hormone plays a role in ovarian cancer pathogenesis.[235] Most of these data derive from studies in the general population, but some information suggests the same is true in women at high risk due to genetic predisposition.

Pregnancy

Among the general population, parity decreases the risk of ovarian cancer by 45% compared with nulliparity. Subsequent pregnancies appear to decrease ovarian cancer risk by 15%.[236] Earlier studies of women with BRCA1/BRCA2 pathogenic variants showed that parity decreases the risk of ovarian cancer.[233,237] In a large case-control study, parity was associated with a significant reduction in ovarian cancer risk in women with BRCA1 pathogenic variants, OR 0.67 (CI, 0.46–0.96).[232] For each birth, carriers of BRCA1 pathogenic variants had an OR of 0.87 (CI, 0.79–0.95). In this same study, parity was associated with an increase in ovarian cancer risk in carriers of BRCA2 pathogenic variants; however, there was no significant trend for each birth, OR 1.08 (CI, 0.90–1.29). Further studies are necessary to define the association of parity and risk of ovarian cancer in carriers of BRCA2 pathogenic variants, but for BRCA1 carriers, each live birth significantly decreases risk of ovarian cancer, as it does in sporadic ovarian cancer.

Lactation and tubal ligation

In the general population, breastfeeding is associated with a decrease in ovarian cancer risk.[238] In carriers of BRCA pathogenic variants, data are limited. One study found no protective effect with breastfeeding.[237] A case-control study among women with BRCA1 or BRCA2 pathogenic variants demonstrates a significant reduction in risk of ovarian cancer (OR, 0.39) for women who have had a tubal ligation. This protective effect was confined to those women with pathogenic variants in BRCA1 and persists after controlling for OC use, parity, history of breast cancer, and ethnicity.[229] A case-control study of ovarian cancer in Israel found a 40% to 50% reduced risk of ovarian cancer among women undergoing gynecologic surgeries (tubal ligation, hysterectomy, unilateral oophorectomy, ovarian cystectomy, excluding bilateral oophorectomy).[172] The mechanism of protection is uncertain. Proposed mechanisms of action include decreased blood flow to the ovary, resulting in interruption of ovulation and/or ovarian hormone production; occlusion of the fallopian tube, thus blocking a pathway for potential carcinogens; or a reduction in the concentration of uterine growth factors that reach the ovary.[239] (Refer to the PDQ summary on Ovarian, Fallopian Tube, and Primary Peritoneal Cancer Prevention for information relevant to the general population.)

Oral contraceptives

Refer to the Oral contraceptives section in the Chemoprevention section of this summary for more information.

Management of Male Carriers of BRCA Pathogenic Variants

There are data to suggest that men with BRCA pathogenic variants have an increased risk of various cancers including male breast cancer and prostate cancer (refer to Table 7).[205,240-244] However, clinical guidelines to manage male carriers with BRCA pathogenic variants are based on consensus statements and expert opinions because information is limited.[33,245,246]

There have been suggestions that BRCA2-associated prostate cancers are associated with aggressive disease phenotype.[247-252] Specifically, two recent studies have reported the median survival of male BRCA2 carriers with prostate cancer in the range of 4 to 5 years.[250,251] Furthermore, mortality rate was reported as 60% at 5 years in one of these studies, compared with 2% to 8% reported in the recent European [253] and North American [254] prostate-specific antigen (PSA) screening trials after comparable follow-up. The data have been more limited in BRCA1-associated prostate cancers, however a number of recent studies have suggested an aggressive disease phenotype as well.[247,249,252,255]

The benefits of PSA screening in BRCA carriers are unknown; however, there have been suggestions (based on very small studies) that PSA levels at prostate cancer diagnosis may be higher in carriers than noncarriers.[256,257] These findings suggest that PSA screening may be of potential utility in men with BRCA pathogenic variants, especially in view of the aggressive phenotype. Preliminary results of the IMPACT PSA screening study reported a PPV of 47.6% in 21 BRCA2 carriers undergoing biopsy on the basis of elevated PSA.[258] Because screening these men detected clinically significant prostate cancer, the authors suggest that these findings provide rationale for continued screening in such men; however, a survival benefit from such screening has not been shown. Ultimately, it is possible that information on BRCA pathogenic variant status in men may inform optimal screening and treatment strategies. Furthermore, recent data that the presence of a germline BRCA2 pathogenic variant is an independent prognostic factor for survival in prostate cancer led these authors to conclude that active surveillance may not be the optimal management strategy due to the aggressive disease phenotype.[251]

NCCN guidelines recommend that males who carry a BRCA pathogenic variant begin breast self-exam training and education at age 35 years. Clinical breast exam is also recommended every 12 months, starting at age 35 years.[33] Annual mammogram screening should be considered in men with gynecomastia starting at age 50 years or 10 years before the earliest known male breast cancer in the family (whichever comes first). Furthermore, beginning at age 40 years, NCCN recommends prostate cancer screening for BRCA2 carriers and the consideration of prostate cancer screening for BRCA1 carriers.[33]

Reproductive Considerations in Carriers of BRCA Pathogenic Variants

Refer to the Prenatal diagnosis and preimplantation genetic testing section in the Psychosocial Issues in BRCA1/BRCA2 section of this summary for more information.

Treatment Strategies

Breast cancer

Prognosis of BRCA1- and BRCA2-related breast cancer
BRCA1-related breast cancer

The distinct features of BRCA1-associated breast tumors are important in prognosis. In addition, there appears to be accelerated growth in BRCA1-associated breast cancer, which is suggested by high-proliferation indices and absence of the expected correlation of tumor size with lymph node status.[259] These pathological features are associated with a worse prognosis in breast cancer, and early studies suggested that carriers of BRCA1 pathogenic variants with breast cancer may have a poorer prognosis compared with sporadic cases.[260-262] These studies particularly noted an increase in ipsilateral and contralateral second primary breast cancers in carriers of BRCA1 and BRCA2 pathogenic variants.[263-267] (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section of this summary for more information.) A retrospective cohort study of 496 Ashkenazi Jewish (AJ) breast cancer patients from two centers compared the relative survival among 56 carriers of BRCA1/BRCA2 pathogenic variants followed up for a median of 116 months. BRCA1 pathogenic variants were independently associated with worse disease-specific survival. The poorer prognosis was not observed in women who received chemotherapy.[268] A large population-based study of incident cases of breast cancer among women in Israel failed to find a difference in OS for carriers of BRCA1 founder pathogenic variants (n = 76) compared with noncarriers (n = 1,189).[269] Similar findings were seen in a European cohort with no differences in disease-free survival in BRCA1-associated breast cancers.[270] A prospective cohort study of 3,220 women from North America and Australia with incident breast cancer (including 93 BRCA1 carriers and 71 BRCA2 carriers) who were followed up for a mean of 7.9 years reported similar outcomes among BRCA1/BRCA2 carriers and those with sporadic disease.[271] However, results were based on chemotherapy regimens used in the late 1990s and did not adjust for surgical approach (lumpectomy vs. mastectomy) and effect of oophorectomy. The Prospective Outcomes in Sporadic versus Hereditary breast cancer (POSH) study recruited 2,733 women, 12% (n = 338) of whom had a BRCA1/BRCA2 pathogenic variant. Carriers showed no significant difference in outcome from noncarriers.[272] However, the cohort of patients with triple-negative breast cancer (n = 558) had a better overall survival than noncarriers at 2 years (HR, 0.59; P = .47), but not a statistically significant difference at 5 and 10 years.

A group of researchers reported the results of BRCA1/BRCA2 testing in 77 unselected patients with triple-negative breast cancer. Of these, 15 (19.5%) had either a germline BRCA1 (n = 11; 14%) or BRCA2 (n = 3; 4%) pathogenic variant or a somatic BRCA1 (n = 1) mutation. The median age at cancer diagnosis was 45 years in carriers of BRCA1 pathogenic variants and 53 years in noncarriers (P = .005). Interestingly, this study also demonstrated a lower risk of relapse in those with triple-negative breast cancer associated with a BRCA1 pathogenic variant than in non-BRCA1-associated triple-negative breast cancer, although this study was limited by its size.[273] Another study examining clinical outcome in BRCA1-associated versus non–BRCA1-associated triple-negative breast cancer showed no difference, although there was a trend toward more brain metastases in those with BRCA1-associated breast cancer. In both of these studies, all but one carrier of a BRCA1 pathogenic variant received chemotherapy.[274] Subsequently, in a study of 89 BRCA1 carriers and 175 noncarriers with triple-negative breast cancer, BRCA1 pathogenic variant status was not an independent predictor of survival after adjusting for age, oophorectomy, and risk-reducing mastectomy.[275] However, carriers who underwent oophorectomy had a significantly lower rate of breast cancer–related death.

A Polish study of 3,345 patients younger than 50 years with stages I through III breast cancer studied the impact of a BRCA1 pathogenic variant on prognosis. In this cohort, 233 patients (7%) carried one of three Polish BRCA1 founder pathogenic variants (5382insC, C61G, or 4154delA). BRCA1 carriers were younger and more frequently ER-negative and HER2/neu-negative. Ten-year survival was similar (80.9% in BRCA1 carriers and 82.2% in noncarriers). Oophorectomy was associated with improved survival in BRCA1 carriers (HR, 0.30; 95% CI 0.12–0.75).[276]

In summary, BRCA1-associated tumors appear to have a prognosis similar to sporadic tumors despite having clinical, histopathologic, and molecular features that indicate a more aggressive phenotype. Carriers of BRCA1 pathogenic variants who do not receive chemotherapy may have a worse prognosis. However, because most BRCA1-associated breast cancers are triple negative, they are usually treated with adjuvant chemotherapy. Work is ongoing to determine whether BRCA1-associated breast cancers should receive different therapy than do sporadic tumors. (Refer to the Role of BRCA1 and BRCA2 in response to systemic therapy section of this summary for more information.)

BRCA2-related breast cancer

Early studies of the prognosis of BRCA2-associated breast cancer have not shown substantial differences in comparison with sporadic breast cancer.[269,277-279] A small study reported statistically significant higher OS in carriers of BRCA2 pathogenic variants with metastatic breast cancer.[270]

Systemic therapy in breast cancer treatment
Role of BRCA1 and BRCA2 in response to systemic therapy
Chemotherapy alone and BRCA pathogenic variants

Retrospective and prospective studies [280-284] have evaluated the response rate to chemotherapy in carriers of BRCA1 pathogenic variants receiving neoadjuvant chemotherapy for breast cancer, especially when using cisplatin.[282] Retrospective reviews have been published detailing pathologic complete response (pCR) rates to standard chemotherapies. In patients treated with standard neoadjuvant anthracycline- and taxane-based chemotherapy regimens, pCR rates ranged from 40% to 60%.[284,285] Neoadjuvant platinum compounds showed impressive pCR rates in patients with BRCA pathogenic variants. A cohort of 107 Polish women with a BRCA1 pathogenic variant received cisplatin 75 mg/m2 every 3 weeks for four cycles; the overall pCR rate was 61%.[286,287] However, in the subset of individuals with known BRCA pathogenic variants in the GeparSixto trial of 50 patients, the pCR rate was 66.7%, but there was no benefit with the addition of a platinum drug to the neoadjuvant chemotherapy regimen that included anthracycline and taxane.[288]

The TNT trial compared docetaxel with carboplatin in 376 patients with metastatic, triple-negative breast cancer. Twenty-nine patients had a pathogenic variant in BRCA1 or BRCA2. There was no difference in the objective response rate (ORR) in the entire cohort between the two arms; however, a difference was noted in the BRCA carriers. The ORR in pathogenic variant carriers who received docetaxel was 33%; the ORR of those patients who received carboplatin was 68% (P = .03).[289]

Targeted therapies with or without chemotherapy

Multiple trials have evaluated the use of poly (ADP-ribose) polymerase (PARP) inhibition with and without chemotherapy. BRCA1 and BRCA2 are active in the repair of double-stranded DNA breaks by homologous recombination; PARP is involved in the repair of single-stranded breaks by base excision repair, as well as by PARP trapping on the DNA strand.

In 2017, two phase III trials explored PARP inhibitors in patients with metastatic breast cancer and a BRCA pathogenic variant. In the OlympiAD trial, 302 patients were randomly assigned to receive olaparib 300 mg orally twice daily or the physician’s choice of chemotherapy (capecitabine, eribulin, or vinorelbine). Progression-free survival (PFS) was improved from a median of 4.2 months to 7.0 months (HR, 0.58; P < .001) in patients treated with olaparib. OS was a secondary endpoint and no statistically significant difference was identified.[290] The EMBRACA trial randomly assigned 431 patients to talazoparib 1 mg orally daily versus the physician’s choice of capecitabine, eribulin, vinorelbine, or gemcitabine.[291] Patients receiving talazoparib had improved PFS by a median of 8.6 months versus 5.6 months (HR, 0.54; P < .001). OS was an alpha-protected endpoint for EMBRACA and, at the time of first report, the data were immature with only 51% of events reported (HR, 0.76; P = .105). Hence, the U.S. Food and Drug Administration has approved the use of both talazoparib and olaparib in patients who have the following: (1) a germline BRCA pathogenic variant and (2) a metastatic breast cancer or an inoperable breast cancer.

PARP inhibitors have also been evaluated in patients with a BRCA pathogenic variant and high-risk, human epidermal growth factor receptor two–negative (HER2-negative), early-stage breast cancers. In the OlympiA trial, 1,836 patients were randomly assigned to receive olaparib or placebo one year after completing local treatment and chemotherapy. Three-hundred milligrams of olaparib was given to patients orally, twice a day.[292] At a planned three-year interim analysis, patients treated with olaparib had significantly increased invasive disease-free survival (DFS) (85.9% vs. 77.1%; HR, 0.58; P < .001) and distant DFS rates (87.5% vs. 80.4%; HR, 0.57; P < .001) than patients who received the placebo. Olaparib was also associated with fewer deaths than the placebo (59 and 86, respectively). However, the between-group difference was not statistically significant.

Ongoing research is evaluating multiple new strategies with PARP inhibitors to include targeting other germline pathogenic variants and somatic mutations. Trials, both in the early and metastatic settings, are evaluating single-agent PARP inhibitors and combining PARP inhibitors with other DNA damage repair agents, immunotherapies, and other targeted therapies to improve responses, as well as broaden the patient population who may benefit.

(Refer to the Systemic therapy in ovarian cancer treatment section in the Ovarian cancer section of this summary for more information about treatment strategies for BRCA-associated ovarian cancer.)

Local therapy
Breast conservation therapy for carriers of BRCA1/BRCA2 pathogenic variants

While lumpectomy plus radiation therapy has become standard local-regional therapy for women with early-stage breast cancer, its use in women with a hereditary predisposition for breast cancer who do not choose immediate bilateral mastectomy is more complicated. Initial concerns about the potential for therapeutic radiation to induce tumors or cause excess toxicity in carriers of BRCA1/BRCA2 pathogenic variants were unfounded.[293-295] Despite this, an increased rate of second primary breast cancer exists, which could impact treatment decisions.

Because of the established increased risk of second primary breast cancers, which may be up to 60% in younger women with BRCA1 pathogenic variants,[265] some carriers of BRCA1/BRCA2 pathogenic variants choose bilateral mastectomy at the time of their initial cancer diagnosis. (Refer to the Contralateral breast cancer in carriers of BRCA pathogenic variants section of this summary for more information.) However, several studies support the use of breast conservation therapy as a reasonable option to treat the primary tumor.[296-298] The risk of ipsilateral recurrence at 10 years has been estimated to be between 10% to 15% and is similar to that seen in noncarriers.[103,265,296-298] Studies with longer periods of follow-up demonstrate risks of ipsilateral breast events at 15 years to be as high as 24%, largely resulting from ipsilateral second breast cancers (rather than relapse of the primary tumor).[296,298,299] In a study of 1,947 consecutive Chinese women treated with breast conservation for primary breast cancer (103 with pathogenic variants in BRCA1/BRCA2 and 1,844 noncarriers), the rate of ipsilateral breast tumor development (new primary or recurrent disease) was 3.9% in pathogenic variant carriers and 2% in noncarriers (P = 0.16). In these cases, however, the new primary rate was significantly higher in those with a pathogenic variant compared with noncarriers (3.9% vs. 0.6%, respectively; P < 0.01).[299] Although not entirely consistent across studies, radiation therapy, chemotherapy, oophorectomy, and tamoxifen are associated with a decreased risk of ipsilateral events,[103,296-298] as is the case in sporadic breast cancer. The risk of contralateral breast cancer does not appear to differ in women undergoing breast conservation therapy versus unilateral mastectomy, suggesting no added risk of contralateral breast cancer from scattered radiation.[296] This finding is supported by a population-based case-control study of women diagnosed with breast cancer before the age of 55 years.[300] All women were genotyped for BRCA1/BRCA2. Although there was a significant fourfold risk of contralateral breast cancer in carriers compared with noncarriers, carriers who were exposed to radiation therapy for the first primary were not at increased risk of contralateral breast cancer compared with carriers who were not exposed. (Refer to the Mammography section for more information about radiation and breast cancer risk.) Finally, no difference in OS at 15 years has been seen between carriers of BRCA1/BRCA2 pathogenic variants choosing breast conservation therapy and carriers choosing mastectomy.[296]

Level of evidence: 3a

Ovarian cancer

Prognosis of BRCA1- and BRCA2-related ovarian cancer

Despite generally poor prognostic factors, several studies have found an improved survival among ovarian cancer patients with BRCA pathogenic variants.[301-309] A nationwide, population-based, case-control study in Israel found 3-year survival rates to be significantly better for ovarian cancer patients with BRCA founder pathogenic variants, compared with controls.[302] Five-year follow-up in the same cohort showed improved survival for carriers of both BRCA1 and BRCA2 pathogenic variants (54 months) versus noncarriers (38 months), which was most pronounced for women with stages III and IV ovarian cancer and for women with high-grade tumors.[310] In a U.S. study of AJ women with ovarian cancer, those with BRCA pathogenic variants had a longer median time to recurrence and an overall improved survival, compared with both AJ women with ovarian cancer who did not have a BRCA pathogenic variant and two large groups of advanced-stage ovarian cancer clinical trial patients.[306] In a retrospective U.S. hospital-based study, AJ carriers of BRCA pathogenic variants had a better response to platinum-based chemotherapy, as measured by response to primary therapy, disease-free survival, and OS, compared with sporadic cases.[304] Similarly, a significant survival advantage was seen in a case-control study among women with non-AJ BRCA pathogenic variants.[311] A study from the Netherlands also showed a better response to platinum-based primary chemotherapy in 112 BRCA1/BRCA2 carriers than in 220 sporadic ovarian cancer patients.[312] A U.S. population-based study showed improvement in OS in BRCA2, but not in BRCA1, carriers.[313] However, the study included only 12 carriers of BRCA2 pathogenic variants and 20 carriers of BRCA1 pathogenic variants. Significantly better OS and PFS were observed in 29 high-grade serous ovarian cancer cases with a known BRCA2 variant (20 germline, 9 somatic) from The Cancer Genome Atlas study compared with cases negative for a BRCA pathogenic variant. BRCA1 pathogenic variants were not significantly associated with prognosis.[314] Furthermore, a pooled analysis of 26 observational studies that included 1,213 carriers of BRCA pathogenic variants and 2,666 noncarriers with epithelial ovarian cancer showed more favorable survival in carriers of pathogenic variants (BRCA1: HR, 0.73; 95% CI, 0.64–0.84; P < .001; BRCA2: HR, 0.49; 95% CI, 0.39–0.61; P < .001).[315] Thus, 5-year survival in both BRCA1 and BRCA2 carriers with epithelial ovarian cancers was better than that observed in noncarriers, with BRCA2 carriers having the best prognosis. A study in Japanese patients found a survival advantage in stage III BRCA1-associated ovarian cancers treated with cisplatin regimens compared with nonhereditary cancers treated in a similar manner.[305]

In contrast, several studies have not found improved OS among ovarian cancer patients with BRCA pathogenic variants.[261,316-318] The largest of these studies involved a large series of unselected Canadian and U.S. patients who were tested for BRCA1 and BRCA2 pathogenic variants. At 3 years, the presence of a pathogenic variant was associated with a better prognosis, but at 10 years, there was no longer a difference seen in prognosis.[319] Furthermore, one study suggested that there was worse survival in ovarian cancer patients with a family history.[317]

Compelling data suggest a short-term survival advantage in carriers of BRCA pathogenic variants. However, long-term outcomes are yet to be established. Survival in AJ ovarian cancer patients with BRCA1 or BRCA2 founder pathogenic variants does seem to be improved;[314,315] however, further large studies in other populations with appropriate controls are needed to determine whether this survival advantage applies more broadly to all BRCA cancers.

Systemic therapy in ovarian cancer treatment

The molecular mechanisms that explain the improved prognosis in hereditary BRCA-associated ovarian cancer are unknown but may be related to the function of BRCA genes. BRCA genes play an important role in cell-cycle checkpoint activation and in the repair of damaged DNA via homologous recombination.[320,321] In addition to BRCA, other genes maintain homologous recombination, such as ATM, BARD1, PALB2, BRIP1, RAD51, BLM, CHEK2, and NBN. Comprehensive genetic testing of larger numbers of ovarian cancers has shown that approximately 50% of serous ovarian tumors may have somatic mutations or germline variants leading to a defective homologous recombination.[322]

Deficiencies in homologous repair can impair the cells’ ability to repair DNA cross-links that result from certain chemotherapy agents, such as cisplatin. Preclinical data has demonstrated BRCA1 impacts chemosensitivity in breast cancer and ovarian cancer cell lines. Reduced BRCA1 protein expression has been shown to enhance cisplatin chemosensitivity.[323] Patients with BRCA-associated ovarian cancer have shown improved responses to both first-line and subsequent platinum-based chemotherapy compared with patients with sporadic cancers, which may contribute to their better outcome.[304,307] Women with ovarian cancer whose tumors have homologous recombination repair gene deficiency (HRD), resulting from either germline variants or somatic mutations, have improved survival compared with women with an intact homologous recombination. The majority of homologous recombination repair gene variants consist of somatic mutations or germline variants in BRCA1 and BRCA2, with one-third contributed by variants in other homologous repair genes.[324,325]

PARP pathway inhibitors have been studied for the treatment of BRCA1- or BRCA2-deficient ovarian cancers. (Refer to the Role of BRCA1 and BRCA2 in response to systemic therapy section in the Treatment Strategies section of this summary for more information about PARP inhibitors.) While PARP is involved in the repair of single-stranded breaks by base excision repair, BRCA1 and BRCA2 are active in the repair of double-stranded DNA breaks by homologous combination. Therefore, it was hypothesized that inhibiting base excision repair with PARP inhibition in BRCA1- or BRCA2-deficient tumors leads to enhanced cell death, as two separate repair mechanisms would be compromised—the concept of synthetic lethality. The same concept may apply to tumors with HRD, and consequently, PARP inhibitors may have expanded use in women whose tumors have any homologous recombination defects beyond pathogenic variants in BRCA genes. In clinical practice, there are different tumor assays available to determine HRD tumors, which vary by method and definition. More study of PARP inhibitors in HRD ovarian cancers is ongoing.

PARP inhibitors
Olaparib

Studies have used PARP inhibitors in ovarian cancer after platinum-based chemotherapy. A phase I study of olaparib, an oral PARP inhibitor, demonstrated tolerability and activity in carriers of BRCA1 and BRCA2 pathogenic variants with ovarian, breast, and prostate cancers.[326] A phase II trial of two different doses of olaparib demonstrated tolerability and efficacy in recurrent ovarian cancer patients with BRCA1 or BRCA2 pathogenic variants.[327] The overall response rate was 33% (11 of 33 patients) in the cohort receiving 400 mg twice daily and 13% (3 of 24 patients) in the cohort receiving 100 mg twice daily (i.e., 16 capsules daily). The most frequent side effects were mild nausea and fatigue.[328] In addition to ovarian cancer patients with germline BRCA1 or BRCA2 pathogenic variants, PARP inhibitors also may be useful in ovarian cancer patients with somatic BRCA1 or BRCA2 mutations or with epigenetic silencing of the genes.[329]

Several phase II treatment studies have explored the efficacy of olaparib in patients with recurrent ovarian cancer, in both platinum-sensitive and platinum-resistant disease. Olaparib at 400 mg twice daily was used in a single-arm study to treat a spectrum of 298 BRCA-associated cancers, including breast, pancreas, prostate, and ovarian. Of the 193 women with ovarian cancer treated with olaparib, 31% had a response, and 40.4% had stable disease that persisted for at least 8 weeks.[330] Among the 154 women previously treated with at least three lines of chemotherapy, a similar overall response rate of 30% was seen, with comparable median durations of response of 8.2 months for platinum-sensitive disease and 8.0 months for platinum-resistant disease.[331] Another study of 173 patients with platinum-sensitive disease were treated with paclitaxel/carboplatin plus olaparib versus paclitaxel/carboplatin alone. The PFS was significantly longer in the olaparib group than the control group (12.2 vs. 9.6 months) (HR, 0.51; 95% CI, 0.34–0.77), especially in the subgroup of patients with BRCA pathogenic variants (HR, 0.21; 95% CI, 0.08–0.55). There were no differences in OS between the olaparib and control groups.[332]

In contrast, other studies found that BRCA status did not predict survival advantage in women with platinum-sensitive ovarian cancer treated with olaparib. A randomized open-label trial assigned 90 women with recurrent platinum-sensitive ovarian cancer to either olaparib or cediranib and olaparib. Median PFS was significantly longer with the combination (17.7 mo vs. 9 mo) (HR, 0.42; 95% CI, 0.23–0.76). Subset analysis showed that combination cediranib and olaparib resulted in significantly longer PFS in the 43 BRCA wild-type/unknown patients than did single agent olaparib (16.5 mo vs. 5.7 mo) (HR, 0.32; P = .008) and a smaller trend toward increased PFS in 47 women with BRCA pathogenic variants (19.4 mo vs. 16.5 mo) (HR, 0.55; P = .16).[333]

In another study, women with BRCA1/BRCA2 pathogenic variants and recurrent ovarian cancer within 12 months of a prior platinum-based regimen were randomly assigned to receive liposomal doxorubicin (Doxil) (n = 33) versus olaparib at 200 mg twice daily (n = 32) versus olaparib at 400 mg twice daily (n = 32). This study did not show a difference in PFS between the groups, which was the primary endpoint.[334] Of interest, the liposomal doxorubicin arm had a higher response rate than anticipated, consistent with other studies demonstrating that BRCA1/BRCA2-associated ovarian cancers may be more sensitive to liposomal doxorubicin than are sporadic ovarian cancers.[335,336] Another study demonstrated significant responses to olaparib in recurrent ovarian cancer patients, including patients with a BRCA1/BRCA2 pathogenic variant (objective response rate [ORR], 41%) and patients without a BRCA1/BRCA2 pathogenic variant (ORR, 24%).[337] This study emphasizes that certain sporadic ovarian cancers, particularly those of high-grade serous histology, may have properties similar to tumors related to a BRCA1/BRCA2 pathogenic variant.

As maintenance treatment, olaparib has shown significantly improved PFS in platinum-sensitive recurrent ovarian cancer. In a randomized controlled study of 265 patients (Study 19), those who received olaparib had a PFS of 8.4 months compared with 4.8 months in those who received the placebo (HR, 0.35; 95% CI, 0.25–0.49).[338] Within the cohort, the 136 patients with BRCA pathogenic variants demonstrated the most benefit with olaparib compared with placebo, with a PFS of 11.2 versus 4.3 months (HR, 0.18; 95% CI, 0.1–0.31).[339] There was no OS difference observed in the entire cohort, or in the carriers of BRCA pathogenic variants. A subsequent post hoc exploratory analysis excluded patients with BRCA pathogenic variants who received a PARP inhibitor at the time of progression to minimize the confounding influence on OS. In this group of 97 patients, an improved OS HR of 0.52 (95% CI, 0.28–0.97) was associated with olaparib, compared with placebo.[340] The more mature Study 19 data, after more than five years of follow-up, showed a trend towards OS benefit but did not meet the a priori significance threshold of P < .0001 with olaparib compared with placebo in the entire cohort (29.8 mo vs. 27.8 mo; HR, 0.73; 95% CI, 0.55–0.96), or among BRCA pathogenic variant carriers treated with olaparib (24.5 mo vs. 26.6 mo; HR, 0.62; 95% CI, 0.41–0.94).[341] Olaparib tablets have been shown to be effective maintenance therapy, compared with placebo, in a similar population of women with recurrent, platinum-sensitive ovarian cancer and BRCA pathogenic variants (SOLO2 trial). Olaparib resulted in a median PFS of 19.1 months versus 5.5 months for placebo (HR, 0.30; 95% CI, 0.22–0.41). Olaparib tablets offer the advantage of a reduced daily pill burden (two tablets twice daily) compared with 16 capsules daily.[342]

Olaparib has demonstrated significant benefit as maintenance treatment in women with newly diagnosed advanced-stage, BRCA-associated ovarian cancer following response to primary treatment. The SOLO-1 trial randomly assigned 391 women with BRCA pathogenic variants to either olaparib 300 mg twice daily (n = 260) or placebo (n = 131) after primary surgery and platinum-based chemotherapy. After a median follow-up of 41 months, women receiving olaparib had a 70% lower risk of disease progression or death compared with women receiving placebo with an estimated improved PFS of approximately 3 years.[343] Within 3 years, disease progression or death occurred in 102 of 260 women (39%) in the olaparib group and 96 of 131 women (73%) in the placebo group. Side effects resulted in a dose reduction in 28% of patients and dose interruptions in more than half of patients. Fatigue and nausea were common side effects and reasons for dose reductions.

Rucaparib

Rucaparib is a small molecule inhibitor of PARP-1, -2, and -3 and was approved in the United States for the treatment of advanced germline BRCA1/BRCA2-associated ovarian cancer in December 2016. A phase II study found that continuous dosing provided better response rates than intermittent dosing in women with pathogenic BRCA-associated breast and ovarian cancer.[344] A subsequent phase I/II dose-finding study selected a dose of 600 mg twice daily on the basis of manageable toxicity and a response rate of 59.5% in 42 women with recurrent, germline BRCA-associated, high-grade serous cancer who had received between two and four prior treatment regimens. Common grade 3 toxicities included fatigue, nausea, and anemia.[345]

The ARIEL-2 phase II study found that rucaparib was effective in the treatment of recurrent, high-grade, platinum-sensitive ovarian cancer in women with BRCA variants, but also in BRCA wild-type women with high genomic loss of heterozygosity (LOH), which is a likely marker of HRD cancers. The study enrolled 206 women, of whom 40 had germline pathogenic variants or somatic mutations in BRCA. An additional 82 were BRCA wild-type but had high LOH. Median PFS was significantly longer in the BRCA variant subgroup (12.8 mo) (HR, 0.27; 95% CI, 0.16–44), and the high LOH subgroup (5.8 mo) (HR, 0.62; 95% CI, 0.42–0.90), compared with the low LOH subgroup (5.2 mo). The authors concluded that both BRCA variant status and LOH score, as a surrogate for HRD, were molecular predictors of rucaparib sensitivity in women with recurrent, platinum-sensitive, high-grade ovarian cancer.[346]

A phase III trial assessed rucaparib versus placebo in 576 women with recurrent, platinum-sensitive, high-grade ovarian cancer after response to second line, or greater, platinum chemotherapy. The study found that 196 women had BRCA pathogenic variants: 130 germline variants and 56 somatic mutations. Median PFS of women in the rucaparib group was 10.8 versus 5.4 months (HR, 0.35; 95% CI, 0.30–0.45). Median PFS was the most prolonged in BRCA-associated ovarian cancer: 16.6 months in the rucaparib group versus 5.4 months in the placebo group (HR, 0.23; 95% CI, 0.16–0.34). In women with HRD cancers, the median PFS was 13.6 versus 5.4 months (HR, 0.32; 95% CI, 0.24–0.42). On the basis of these data, the authors concluded that platinum sensitivity alone was a sufficient marker to predict benefit from rucaparib in women with advanced high-grade ovarian cancer, without requiring additional HRD or BRCA testing.[347]

Niraparib

Niraparib is a selective inhibitor of PARP-1 and -2. A phase I dose-finding study observed a response rate of 42% with 300 mg daily in women with recurrent, BRCA-associated solid tumors.[348] In a cohort of 500 patients with platinum-sensitive, recurrent ovarian cancer, 234 received niraparib maintenance treatment and 116 received placebo (NOVA trial).[349] Niraparib maintenance resulted in improved PFS in BRCA pathogenic variant carriers (at 21 mo) and in wild-type patients with HRD positivity (at 12 mo) compared with wild-type patients without HRD tumor positivity (at 9 mo). Consistent with prior data, patients with germline BRCA pathogenic variants had the longest PFS of the three groups. Based upon the broad activity of niraparib maintenance in heavily pretreated women with ovarian cancer, regardless of platinum response or variant status, the QUADRA phase II trial studied the antitumor activity of niraparib in 463 women with recurrent, measurable ovarian cancer. Women had received a median of four prior lines of treatment. Twenty-eight percent of women had an overall response with a median duration of 9 months, which was improved in platinum sensitive, HRD-positive women.[350]

More mature data are necessary to determine whether platinum sensitivity alone is a marker of response to PARP inhibitors in women with BRCA pathogenic variants, and the optimal timing of PARP inhibitors as treatment or as maintenance therapy. HRD status may also be used to predict response to PARP treatment on the basis of a better understanding of the multiple genes involved in homologous repair pathways.

Level of evidence: 3dii

Available Clinical Practice Guidelines for Hereditary Breast and Ovarian Cancer

Table 13 lists several organizations that have published recommendations for cancer risk assessment and genetic counseling, genetic testing, and/or management for hereditary breast and ovarian cancer.

Table 13. Available Clinical Practice Guidelines for Hereditary Breast and Ovarian Cancer (HBOC)
Organization Referral Recommendations Risk Assessment and Genetic Counseling Recommendations Genetic Testing Recommendations Management Recommendations
ACMG/NSGC = American College of Medical Genetics and Genomics/National Society of Genetic Counselors; ACOG = American College of Obstetricians and Gynecologists; ASCO = American Society of Clinical Oncology; ESMO = European Society for Medical Oncology; NAPBC = National Accreditation Program for Breast Centers; NCCN = National Comprehensive Cancer Network; NSGC = National Society of Genetic Counselors; SGO = Society of Gynecologic Oncology; USPSTF = U.S. Preventive Services Task Force.
aThe USPSTF guidelines apply to individuals without a prior cancer diagnosis.
ACMG/NSGC (2015) [351] Addressed Risk Assessment: Addressed Not addressed Not addressed
Genetic Counseling: Addressed
ACOG (2017) [352] Addressed Risk Assessment: Addressed Addressed Addressed
Genetic Counseling: Addressed
ASCO (2015) [353] Not addressed Risk Assessment: General recommendations; not specific to HBOC General recommendations; not specific to HBOC Not addressed
Genetic Counseling: Addressed
ESMO (2016) [354] Refers to other published guidelines Risk Assessment: Refers to other published guidelines Refers to other published guidelines Addressed
Genetic Counseling: Addressed
NAPBC (2014) [355] Refers to other published guidelines Risk Assessment: Refers to other published guidelines Indications for testing not addressed; components of pretest and posttest counseling addressed Not addressed
Genetic Counseling: Addressed
NSGC (2013) [356] Addressed Risk Assessment: Refers to other published guidelines and available models Addressed Refers to other published guidelines
Genetic Counseling: Addressed
NCCN (2022) [33] Addressed Risk Assessment: Addressed Addressed Addressed
Genetic Counseling: Addressed
SGO (2015, 2017) [352,357] Addressed Risk Assessment: Addressed Addressed Addressed
Genetic Counseling: Addressed
USPSTFa (2019) [358] Addressed Risk Assessment: Addressed Addressed in general terms and other guidelines referenced Addressed in general terms and other guidelines referenced
Genetic Counseling: Addressed
References
  1. U.S. Preventive Services Task Force: Genetic risk assessment and BRCA mutation testing for breast and ovarian cancer susceptibility: recommendation statement. Ann Intern Med 143 (5): 355-61, 2005. [PUBMED Abstract]
  2. 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 304 (9): 967-75, 2010. [PUBMED Abstract]
  3. 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 22 (6): 1055-62, 2004. [PUBMED Abstract]
  4. Evans DG, Ingham SL, Baildam A, et al.: Contralateral mastectomy improves survival in women with BRCA1/2-associated breast cancer. Breast Cancer Res Treat 140 (1): 135-42, 2013. [PUBMED Abstract]
  5. Kurian AW, Sigal BM, Plevritis SK: Survival analysis of cancer risk reduction strategies for BRCA1/2 mutation carriers. J Clin Oncol 28 (2): 222-31, 2010. [PUBMED Abstract]
  6. Kauff ND, Domchek SM, Friebel TM, et al.: Risk-reducing salpingo-oophorectomy for the prevention of BRCA1- and BRCA2-associated breast and gynecologic cancer: a multicenter, prospective study. J Clin Oncol 26 (8): 1331-7, 2008. [PUBMED Abstract]
  7. Finch AP, Lubinski J, Møller P, et al.: Impact of oophorectomy on cancer incidence and mortality in women with a BRCA1 or BRCA2 mutation. J Clin Oncol 32 (15): 1547-53, 2014. [PUBMED Abstract]
  8. Thomas DB, Gao DL, Self SG, et al.: Randomized trial of breast self-examination in Shanghai: methodology and preliminary results. J Natl Cancer Inst 89 (5): 355-65, 1997. [PUBMED Abstract]
  9. 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 20 (5): 1260-8, 2002. [PUBMED Abstract]
  10. 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 19 (4): 924-30, 2001. [PUBMED Abstract]
  11. Burke W, Daly M, Garber J, et al.: Recommendations for follow-up care of individuals with an inherited predisposition to cancer. II. BRCA1 and BRCA2. Cancer Genetics Studies Consortium. JAMA 277 (12): 997-1003, 1997. [PUBMED Abstract]
  12. Shapiro S: Periodic screening for breast cancer: the Health Insurance Plan project and its sequelae, 1963-1986. Johns Hopkins University Press, 1988.
  13. Kerlikowske K, Grady D, Barclay J, et al.: Effect of age, breast density, and family history on the sensitivity of first screening mammography. JAMA 276 (1): 33-8, 1996. [PUBMED Abstract]
  14. Kerlikowske K, Carney PA, Geller B, et al.: Performance of screening mammography among women with and without a first-degree relative with breast cancer. Ann Intern Med 133 (11): 855-63, 2000. [PUBMED Abstract]
  15. Kerlikowske K, Grady D, Barclay J, et al.: Positive predictive value of screening mammography by age and family history of breast cancer. JAMA 270 (20): 2444-50, 1993. [PUBMED Abstract]
  16. Tilanus-Linthorst M, Verhoog L, Obdeijn IM, et al.: A BRCA1/2 mutation, high breast density and prominent pushing margins of a tumor independently contribute to a frequent false-negative mammography. Int J Cancer 102 (1): 91-5, 2002. [PUBMED Abstract]
  17. Tilanus-Linthorst MM, Kriege M, Boetes C, et al.: Hereditary breast cancer growth rates and its impact on screening policy. Eur J Cancer 41 (11): 1610-7, 2005. [PUBMED Abstract]
  18. Mitchell G, Antoniou AC, Warren R, et al.: Mammographic density and breast cancer risk in BRCA1 and BRCA2 mutation carriers. Cancer Res 66 (3): 1866-72, 2006. [PUBMED Abstract]
  19. Miller AB, To T, Baines CJ, et al.: Canadian National Breast Screening Study-2: 13-year results of a randomized trial in women aged 50-59 years. J Natl Cancer Inst 92 (18): 1490-9, 2000. [PUBMED Abstract]
  20. Shtern F: Digital mammography and related technologies: a perspective from the National Cancer Institute. Radiology 183 (3): 629-30, 1992. [PUBMED Abstract]
  21. Lewin JM, D'Orsi CJ, Hendrick RE, et al.: Clinical comparison of full-field digital mammography and screen-film mammography for detection of breast cancer. AJR Am J Roentgenol 179 (3): 671-7, 2002. [PUBMED Abstract]
  22. Pisano ED, Gatsonis C, Hendrick E, et al.: Diagnostic performance of digital versus film mammography for breast-cancer screening. N Engl J Med 353 (17): 1773-83, 2005. [PUBMED Abstract]
  23. Sharan SK, Morimatsu M, Albrecht U, et al.: Embryonic lethality and radiation hypersensitivity mediated by Rad51 in mice lacking Brca2. Nature 386 (6627): 804-10, 1997. [PUBMED Abstract]
  24. Gowen LC, Avrutskaya AV, Latour AM, et al.: BRCA1 required for transcription-coupled repair of oxidative DNA damage. Science 281 (5379): 1009-12, 1998. [PUBMED Abstract]
  25. Abbott DW, Freeman ML, Holt JT: Double-strand break repair deficiency and radiation sensitivity in BRCA2 mutant cancer cells. J Natl Cancer Inst 90 (13): 978-85, 1998. [PUBMED Abstract]
  26. 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 7 (5): 402-6, 2006. [PUBMED Abstract]
  27. Goldfrank D, Chuai S, Bernstein JL, et al.: Effect of mammography on breast cancer risk in women with mutations in BRCA1 or BRCA2. Cancer Epidemiol Biomarkers Prev 15 (11): 2311-3, 2006. [PUBMED Abstract]
  28. Giannakeas V, Lubinski J, Gronwald J, et al.: Mammography screening and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers: a prospective study. Breast Cancer Res Treat 147 (1): 113-8, 2014. [PUBMED Abstract]
  29. 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 24 (21): 3361-6, 2006. [PUBMED Abstract]
  30. 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 345: e5660, 2012. [PUBMED Abstract]
  31. Berrington de Gonzalez A, Berg CD, Visvanathan K, et al.: Estimated risk of radiation-induced breast cancer from mammographic screening for young BRCA mutation carriers. J Natl Cancer Inst 101 (3): 205-9, 2009. [PUBMED Abstract]
  32. Lowry KP, Lee JM, Kong CY, et al.: Annual screening strategies in BRCA1 and BRCA2 gene mutation carriers: a comparative effectiveness analysis. Cancer 118 (8): 2021-30, 2012. [PUBMED Abstract]
  33. National Comprehensive Cancer Network: NCCN Clinical Practice Guidelines in Oncology: Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 2.2022. Plymouth Meeting, Pa: National Comprehensive Cancer Network, 2022. Available online with free registration. Last accessed March 16, 2022.
  34. 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 351 (5): 427-37, 2004. [PUBMED Abstract]
  35. Lehman CD, Blume JD, Weatherall P, et al.: Screening women at high risk for breast cancer with mammography and magnetic resonance imaging. Cancer 103 (9): 1898-905, 2005. [PUBMED Abstract]
  36. Leach MO, Boggis CR, Dixon AK, et al.: Screening with magnetic resonance imaging and mammography of a UK population at high familial risk of breast cancer: a prospective multicentre cohort study (MARIBS). Lancet 365 (9473): 1769-78, 2005 May 21-27. [PUBMED Abstract]
  37. Warner E, Plewes DB, Hill KA, et al.: Surveillance of BRCA1 and BRCA2 mutation carriers with magnetic resonance imaging, ultrasound, mammography, and clinical breast examination. JAMA 292 (11): 1317-25, 2004. [PUBMED Abstract]
  38. 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 244 (2): 381-8, 2007. [PUBMED Abstract]
  39. Sardanelli F, Podo F, D'Agnolo G, et al.: Multicenter comparative multimodality surveillance of women at genetic-familial high risk for breast cancer (HIBCRIT study): interim results. Radiology 242 (3): 698-715, 2007. [PUBMED Abstract]
  40. Kuhl C, Weigel S, Schrading S, et al.: Prospective multicenter cohort study to refine management recommendations for women at elevated familial risk of breast cancer: the EVA trial. J Clin Oncol 28 (9): 1450-7, 2010. [PUBMED Abstract]
  41. Shah P, Rosen M, Stopfer J, et al.: Prospective study of breast MRI in BRCA1 and BRCA2 mutation carriers: effect of mutation status on cancer incidence. Breast Cancer Res Treat 118 (3): 539-46, 2009. [PUBMED Abstract]
  42. Rijnsburger AJ, Obdeijn IM, Kaas R, et al.: BRCA1-associated breast cancers present differently from BRCA2-associated and familial cases: long-term follow-up of the Dutch MRISC Screening Study. J Clin Oncol 28 (36): 5265-73, 2010. [PUBMED Abstract]
  43. Weinstein SP, Localio AR, Conant EF, et al.: Multimodality screening of high-risk women: a prospective cohort study. J Clin Oncol 27 (36): 6124-8, 2009. [PUBMED Abstract]
  44. 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 46 (2): 94-105, 2011. [PUBMED Abstract]
  45. Lord SJ, Lei W, Craft P, et al.: A systematic review of the effectiveness of magnetic resonance imaging (MRI) as an addition to mammography and ultrasound in screening young women at high risk of breast cancer. Eur J Cancer 43 (13): 1905-17, 2007. [PUBMED Abstract]
  46. Obdeijn IM, Loo CE, Rijnsburger AJ, et al.: Assessment of false-negative cases of breast MR imaging in women with a familial or genetic predisposition. Breast Cancer Res Treat 119 (2): 399-407, 2010. [PUBMED Abstract]
  47. Saadatmand S, Geuzinge HA, Rutgers EJT, et al.: MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): a multicentre, randomised, controlled trial. Lancet Oncol 20 (8): 1136-1147, 2019. [PUBMED Abstract]
  48. Heijnsdijk EA, Warner E, Gilbert FJ, et al.: Differences in natural history between breast cancers in BRCA1 and BRCA2 mutation carriers and effects of MRI screening-MRISC, MARIBS, and Canadian studies combined. Cancer Epidemiol Biomarkers Prev 21 (9): 1458-68, 2012. [PUBMED Abstract]
  49. Passaperuma K, Warner E, Causer PA, et al.: Long-term results of screening with magnetic resonance imaging in women with BRCA mutations. Br J Cancer 107 (1): 24-30, 2012. [PUBMED Abstract]
  50. Saadatmand S, Obdeijn IM, Rutgers EJ, et al.: Survival benefit in women with BRCA1 mutation or familial risk in the MRI screening study (MRISC). Int J Cancer 137 (7): 1729-38, 2015. [PUBMED Abstract]
  51. 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 57 (2): 75-89, 2007 Mar-Apr. [PUBMED Abstract]
  52. Golan O, Amitai Y, Barnea Y, et al.: Yield of surveillance magnetic resonance imaging after bilateral mastectomy and reconstruction: a retrospective cohort study. Breast Cancer Res Treat 174 (2): 463-468, 2019. [PUBMED Abstract]
  53. O'Driscoll D, Warren R, MacKay J, et al.: Screening with breast ultrasound in a population at moderate risk due to family history. J Med Screen 8 (2): 106-9, 2001. [PUBMED Abstract]
  54. Berg WA, Blume JD, Cormack JB, et al.: Combined screening with ultrasound and mammography vs mammography alone in women at elevated risk of breast cancer. JAMA 299 (18): 2151-63, 2008. [PUBMED Abstract]
  55. Giuliano AE, Boolbol S, Degnim A, et al.: Society of Surgical Oncology: position statement on prophylactic mastectomy. Approved by the Society of Surgical Oncology Executive Council, March 2007. Ann Surg Oncol 14 (9): 2425-7, 2007. [PUBMED Abstract]
  56. Hartmann LC, Sellers TA, Schaid DJ, et al.: Efficacy of bilateral prophylactic mastectomy in BRCA1 and BRCA2 gene mutation carriers. J Natl Cancer Inst 93 (21): 1633-7, 2001. [PUBMED Abstract]
  57. 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 345 (3): 159-64, 2001. [PUBMED Abstract]
  58. 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 340 (2): 77-84, 1999. [PUBMED Abstract]
  59. Schrag D, Kuntz KM, Garber JE, et al.: Decision analysis--effects of prophylactic mastectomy and oophorectomy on life expectancy among women with BRCA1 or BRCA2 mutations. N Engl J Med 336 (20): 1465-71, 1997. [PUBMED Abstract]
  60. Kurian AW, Munoz DF, Rust P, et al.: Online tool to guide decisions for BRCA1/2 mutation carriers. J Clin Oncol 30 (5): 497-506, 2012. [PUBMED Abstract]
  61. Unic I, Stalmeier PF, Verhoef LC, et al.: Assessment of the time-tradeoff values for prophylactic mastectomy of women with a suspected genetic predisposition to breast cancer. Med Decis Making 18 (3): 268-77, 1998 Jul-Sep. [PUBMED Abstract]
  62. Grann VR, Panageas KS, Whang W, et al.: Decision analysis of prophylactic mastectomy and oophorectomy in BRCA1-positive or BRCA2-positive patients. J Clin Oncol 16 (3): 979-85, 1998. [PUBMED Abstract]
  63. Tuttle TM, Habermann EB, Grund EH, et al.: Increasing use of contralateral prophylactic mastectomy for breast cancer patients: a trend toward more aggressive surgical treatment. J Clin Oncol 25 (33): 5203-9, 2007. [PUBMED Abstract]
  64. Jones NB, Wilson J, Kotur L, et al.: Contralateral prophylactic mastectomy for unilateral breast cancer: an increasing trend at a single institution. Ann Surg Oncol 16 (10): 2691-6, 2009. [PUBMED Abstract]
  65. Nichols HB, Berrington de González A, Lacey JV, et al.: Declining incidence of contralateral breast cancer in the United States from 1975 to 2006. J Clin Oncol 29 (12): 1564-9, 2011. [PUBMED Abstract]
  66. Fayanju OM, Stoll CR, Fowler S, et al.: Contralateral prophylactic mastectomy after unilateral breast cancer: a systematic review and meta-analysis. Ann Surg 260 (6): 1000-10, 2014. [PUBMED Abstract]
  67. Metcalfe K, Gershman S, Ghadirian P, et al.: Contralateral mastectomy and survival after breast cancer in carriers of BRCA1 and BRCA2 mutations: retrospective analysis. BMJ 348: g226, 2014. [PUBMED Abstract]
  68. Chiba A, Hoskin TL, Hallberg EJ, et al.: Impact that Timing of Genetic Mutation Diagnosis has on Surgical Decision Making and Outcome for BRCA1/BRCA2 Mutation Carriers with Breast Cancer. Ann Surg Oncol 23 (10): 3232-8, 2016. [PUBMED Abstract]
  69. van Sprundel TC, Schmidt MK, Rookus MA, et al.: Risk reduction of contralateral breast cancer and survival after contralateral prophylactic mastectomy in BRCA1 or BRCA2 mutation carriers. Br J Cancer 93 (3): 287-92, 2005. [PUBMED Abstract]
  70. Evans DG, Baildam AD, Anderson E, et al.: Risk reducing mastectomy: outcomes in 10 European centres. J Med Genet 46 (4): 254-8, 2009. [PUBMED Abstract]
  71. Heemskerk-Gerritsen BA, Rookus MA, Aalfs CM, et al.: Improved overall survival after contralateral risk-reducing mastectomy in BRCA1/2 mutation carriers with a history of unilateral breast cancer: a prospective analysis. Int J Cancer 136 (3): 668-77, 2015. [PUBMED Abstract]
  72. Jatoi I, Parsons HM: Contralateral prophylactic mastectomy and its association with reduced mortality: evidence for selection bias. Breast Cancer Res Treat 148 (2): 389-96, 2014. [PUBMED Abstract]
  73. Yao K, Liederbach E, Tang R, et al.: Nipple-sparing mastectomy in BRCA1/2 mutation carriers: an interim analysis and review of the literature. Ann Surg Oncol 22 (2): 370-6, 2015. [PUBMED Abstract]
  74. Manning AT, Wood C, Eaton A, et al.: Nipple-sparing mastectomy in patients with BRCA1/2 mutations and variants of uncertain significance. Br J Surg 102 (11): 1354-9, 2015. [PUBMED Abstract]
  75. 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 153 (2): 123-129, 2018. [PUBMED Abstract]
  76. Kauff ND, Brogi E, Scheuer L, et al.: Epithelial lesions in prophylactic mastectomy specimens from women with BRCA mutations. Cancer 97 (7): 1601-8, 2003. [PUBMED Abstract]
  77. Hoogerbrugge N, Bult P, de Widt-Levert LM, et al.: High prevalence of premalignant lesions in prophylactically removed breasts from women at hereditary risk for breast cancer. J Clin Oncol 21 (1): 41-5, 2003. [PUBMED Abstract]
  78. Kroiss R, Winkler V, Kalteis K, et al.: Prevalence of pre-malignant and malignant lesions in prophylactic mastectomy specimens of BRCA1 mutation carriers: comparison with a control group. J Cancer Res Clin Oncol 134 (10): 1113-21, 2008. [PUBMED Abstract]
  79. Scott CI, Iorgulescu DG, Thorne HJ, et al.: Clinical, pathological and genetic features of women at high familial risk of breast cancer undergoing prophylactic mastectomy. Clin Genet 64 (2): 111-21, 2003. [PUBMED Abstract]
  80. Olson JE, Sellers TA, Iturria SJ, et al.: Bilateral oophorectomy and breast cancer risk reduction among women with a family history. Cancer Detect Prev 28 (5): 357-60, 2004. [PUBMED Abstract]
  81. Rebbeck TR, Lynch HT, Neuhausen SL, et al.: Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. N Engl J Med 346 (21): 1616-22, 2002. [PUBMED Abstract]
  82. Struewing JP, Watson P, Easton DF, et al.: Prophylactic oophorectomy in inherited breast/ovarian cancer families. J Natl Cancer Inst Monogr (17): 33-5, 1995. [PUBMED Abstract]
  83. Rebbeck TR, Levin AM, Eisen A, et al.: Breast cancer risk after bilateral prophylactic oophorectomy in BRCA1 mutation carriers. J Natl Cancer Inst 91 (17): 1475-9, 1999. [PUBMED Abstract]
  84. Kauff ND, Satagopan JM, Robson ME, et al.: Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346 (21): 1609-15, 2002. [PUBMED Abstract]
  85. Rebbeck TR, Kauff ND, Domchek SM: Meta-analysis of risk reduction estimates associated with risk-reducing salpingo-oophorectomy in BRCA1 or BRCA2 mutation carriers. J Natl Cancer Inst 101 (2): 80-7, 2009. [PUBMED Abstract]
  86. Heemskerk-Gerritsen BA, Seynaeve C, van Asperen CJ, et al.: Breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: revisiting the evidence for risk reduction. J Natl Cancer Inst 107 (5): , 2015. [PUBMED Abstract]
  87. Chai X, Domchek S, Kauff N, et al.: RE: Breast Cancer Risk After Salpingo-Oophorectomy in Healthy BRCA1/2 Mutation Carriers: Revisiting the Evidence for Risk Reduction. J Natl Cancer Inst 107 (9): , 2015. [PUBMED Abstract]
  88. Metcalfe K, Lynch HT, Foulkes WD, et al.: Effect of Oophorectomy on Survival After Breast Cancer in BRCA1 and BRCA2 Mutation Carriers. JAMA Oncol 1 (3): 306-13, 2015. [PUBMED Abstract]
  89. Kotsopoulos J, Huzarski T, Gronwald J, et al.: Bilateral Oophorectomy and Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers. J Natl Cancer Inst 109 (1): , 2017. [PUBMED Abstract]
  90. Terry MB, Daly MB, Phillips KA, et al.: Risk-Reducing Oophorectomy and Breast Cancer Risk Across the Spectrum of Familial Risk. J Natl Cancer Inst 111 (3): 331-334, 2019. [PUBMED Abstract]
  91. 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 14: 150, 2014. [PUBMED Abstract]
  92. Fisher B, Costantino JP, Wickerham DL, et al.: Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 90 (18): 1371-88, 1998. [PUBMED Abstract]
  93. Veronesi U, Maisonneuve P, Costa A, et al.: Prevention of breast cancer with tamoxifen: preliminary findings from the Italian randomised trial among hysterectomised women. Italian Tamoxifen Prevention Study. Lancet 352 (9122): 93-7, 1998. [PUBMED Abstract]
  94. Powles T, Eeles R, Ashley S, et al.: Interim analysis of the incidence of breast cancer in the Royal Marsden Hospital tamoxifen randomised chemoprevention trial. Lancet 352 (9122): 98-101, 1998. [PUBMED Abstract]
  95. Cuzick J, Sestak I, Cawthorn S, et al.: Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol 16 (1): 67-75, 2015. [PUBMED Abstract]
  96. 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 286 (18): 2251-6, 2001. [PUBMED Abstract]
  97. Narod SA, Brunet JS, Ghadirian P, et al.: Tamoxifen and risk of contralateral breast cancer in BRCA1 and BRCA2 mutation carriers: a case-control study. Hereditary Breast Cancer Clinical Study Group. Lancet 356 (9245): 1876-81, 2000. [PUBMED Abstract]
  98. Pierce LJ, Levin AM, Rebbeck TR, et al.: Ten-year multi-institutional results of breast-conserving surgery and radiotherapy in BRCA1/2-associated stage I/II breast cancer. J Clin Oncol 24 (16): 2437-43, 2006. [PUBMED Abstract]
  99. Gronwald J, Tung N, Foulkes WD, et al.: Tamoxifen and contralateral breast cancer in BRCA1 and BRCA2 carriers: an update. Int J Cancer 118 (9): 2281-4, 2006. [PUBMED Abstract]
  100. Phillips KA, Milne RL, Rookus MA, et al.: Tamoxifen and risk of contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. J Clin Oncol 31 (25): 3091-9, 2013. [PUBMED Abstract]
  101. 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 295 (23): 2727-41, 2006. [PUBMED Abstract]
  102. Land SR, Wickerham DL, Costantino JP, et al.: Patient-reported symptoms and quality of life during treatment with tamoxifen or raloxifene for breast cancer prevention: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA 295 (23): 2742-51, 2006. [PUBMED Abstract]
  103. Reding KW, Bernstein JL, Langholz BM, et al.: Adjuvant systemic therapy for breast cancer in BRCA1/BRCA2 mutation carriers in a population-based study of risk of contralateral breast cancer. Breast Cancer Res Treat 123 (2): 491-8, 2010. [PUBMED Abstract]
  104. Vicus D, Rosen B, Lubinski J, et al.: Tamoxifen and the risk of ovarian cancer in BRCA1 mutation carriers. Gynecol Oncol 115 (1): 135-7, 2009. [PUBMED Abstract]
  105. Colditz GA, Rosner BA, Speizer FE: Risk factors for breast cancer according to family history of breast cancer. For the Nurses' Health Study Research Group. J Natl Cancer Inst 88 (6): 365-71, 1996. [PUBMED Abstract]
  106. Narod S, Lynch H, Conway T, et al.: Increasing incidence of breast cancer in family with BRCA1 mutation. Lancet 341 (8852): 1101-2, 1993. [PUBMED Abstract]
  107. Narod SA, Goldgar D, Cannon-Albright L, et al.: Risk modifiers in carriers of BRCA1 mutations. Int J Cancer 64 (6): 394-8, 1995. [PUBMED Abstract]
  108. McCredie M, Paul C, Skegg DC, et al.: Family history and risk of breast cancer in New Zealand. Int J Cancer 73 (4): 503-7, 1997. [PUBMED Abstract]
  109. Jernström H, Lerman C, Ghadirian P, et al.: Pregnancy and risk of early breast cancer in carriers of BRCA1 and BRCA2. Lancet 354 (9193): 1846-50, 1999. [PUBMED Abstract]
  110. Cullinane CA, Lubinski J, Neuhausen SL, et al.: Effect of pregnancy as a risk factor for breast cancer in BRCA1/BRCA2 mutation carriers. Int J Cancer 117 (6): 988-91, 2005. [PUBMED Abstract]
  111. Friedman E, Kotsopoulos J, Lubinski J, et al.: Spontaneous and therapeutic abortions and the risk of breast cancer among BRCA mutation carriers. Breast Cancer Res 8 (2): R15, 2006. [PUBMED Abstract]
  112. Milne RL, Osorio A, Ramón y Cajal T, et al.: Parity and the risk of breast and ovarian cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 119 (1): 221-32, 2010. [PUBMED Abstract]
  113. Antoniou AC, Shenton A, Maher ER, et al.: Parity and breast cancer risk among BRCA1 and BRCA2 mutation carriers. Breast Cancer Res 8 (6): R72, 2006. [PUBMED Abstract]
  114. Andrieu N, Goldgar DE, Easton DF, et al.: Pregnancies, breast-feeding, and breast cancer risk in the International BRCA1/2 Carrier Cohort Study (IBCCS). J Natl Cancer Inst 98 (8): 535-44, 2006. [PUBMED Abstract]
  115. Col: Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet 360 (9328): 187-95, 2002. [PUBMED Abstract]
  116. Jernström H, Lubinski J, Lynch HT, et al.: Breast-feeding and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 96 (14): 1094-8, 2004. [PUBMED Abstract]
  117. Breast cancer and hormonal contraceptives: collaborative reanalysis of individual data on 53 297 women with breast cancer and 100 239 women without breast cancer from 54 epidemiological studies. Collaborative Group on Hormonal Factors in Breast Cancer. Lancet 347 (9017): 1713-27, 1996. [PUBMED Abstract]
  118. Ursin G, Henderson BE, Haile RW, et al.: Does oral contraceptive use increase the risk of breast cancer in women with BRCA1/BRCA2 mutations more than in other women? Cancer Res 57 (17): 3678-81, 1997. [PUBMED Abstract]
  119. Jernström H, Loman N, Johannsson OT, et al.: Impact of teenage oral contraceptive use in a population-based series of early-onset breast cancer cases who have undergone BRCA mutation testing. Eur J Cancer 41 (15): 2312-20, 2005. [PUBMED Abstract]
  120. 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 46 (12): 2275-84, 2010. [PUBMED Abstract]
  121. Brohet RM, Goldgar DE, Easton DF, et al.: Oral contraceptives and breast cancer risk in the international BRCA1/2 carrier cohort study: a report from EMBRACE, GENEPSO, GEO-HEBON, and the IBCCS Collaborating Group. J Clin Oncol 25 (25): 3831-6, 2007. [PUBMED Abstract]
  122. Haile RW, Thomas DC, McGuire V, et al.: BRCA1 and BRCA2 mutation carriers, oral contraceptive use, and breast cancer before age 50. Cancer Epidemiol Biomarkers Prev 15 (10): 1863-70, 2006. [PUBMED Abstract]
  123. Narod SA, Dubé MP, Klijn J, et al.: Oral contraceptives and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. J Natl Cancer Inst 94 (23): 1773-9, 2002. [PUBMED Abstract]
  124. Kotsopoulos J, Lubinski J, Moller P, et al.: Timing of oral contraceptive use and the risk of breast cancer in BRCA1 mutation carriers. Breast Cancer Res Treat 143 (3): 579-86, 2014. [PUBMED Abstract]
  125. Narod SA, Risch H, Moslehi R, et al.: Oral contraceptives and the risk of hereditary ovarian cancer. Hereditary Ovarian Cancer Clinical Study Group. N Engl J Med 339 (7): 424-8, 1998. [PUBMED Abstract]
  126. Chen CL, Weiss NS, Newcomb P, et al.: Hormone replacement therapy in relation to breast cancer. JAMA 287 (6): 734-41, 2002. [PUBMED Abstract]
  127. Writing Group for the Women's Health Initiative Investigators: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA 288 (3): 321-33, 2002. [PUBMED Abstract]
  128. Chlebowski RT, Hendrix SL, Langer RD, et al.: Influence of estrogen plus progestin on breast cancer and mammography in healthy postmenopausal women: the Women's Health Initiative Randomized Trial. JAMA 289 (24): 3243-53, 2003. [PUBMED Abstract]
  129. Chlebowski RT, Kuller LH, Prentice RL, et al.: Breast cancer after use of estrogen plus progestin in postmenopausal women. N Engl J Med 360 (6): 573-87, 2009. [PUBMED Abstract]
  130. Schuurman AG, van den Brandt PA, Goldbohm RA: Exogenous hormone use and the risk of postmenopausal breast cancer: results from The Netherlands Cohort Study. Cancer Causes Control 6 (5): 416-24, 1995. [PUBMED Abstract]
  131. Steinberg KK, Thacker SB, Smith SJ, et al.: A meta-analysis of the effect of estrogen replacement therapy on the risk of breast cancer. JAMA 265 (15): 1985-90, 1991. [PUBMED Abstract]
  132. Colditz GA, Egan KM, Stampfer MJ: Hormone replacement therapy and risk of breast cancer: results from epidemiologic studies. Am J Obstet Gynecol 168 (5): 1473-80, 1993. [PUBMED Abstract]
  133. Sellers TA, Mink PJ, Cerhan JR, et al.: The role of hormone replacement therapy in the risk for breast cancer and total mortality in women with a family history of breast cancer. Ann Intern Med 127 (11): 973-80, 1997. [PUBMED Abstract]
  134. Stanford JL, Weiss NS, Voigt LF, et al.: Combined estrogen and progestin hormone replacement therapy in relation to risk of breast cancer in middle-aged women. JAMA 274 (2): 137-42, 1995. [PUBMED Abstract]
  135. Gorsky RD, Koplan JP, Peterson HB, et al.: Relative risks and benefits of long-term estrogen replacement therapy: a decision analysis. Obstet Gynecol 83 (2): 161-6, 1994. [PUBMED Abstract]
  136. 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 23 (31): 7804-10, 2005. [PUBMED Abstract]
  137. Eisen A, Lubinski J, Gronwald J, et al.: Hormone therapy and the risk of breast cancer in BRCA1 mutation carriers. J Natl Cancer Inst 100 (19): 1361-7, 2008. [PUBMED Abstract]
  138. Kotsopoulos J, Huzarski T, Gronwald J, et al.: Hormone replacement therapy after menopause and risk of breast cancer in BRCA1 mutation carriers: a case-control study. Breast Cancer Res Treat 155 (2): 365-73, 2016. [PUBMED Abstract]
  139. Chlebowski RT, Prentice RL: Menopausal hormone therapy in BRCA1 mutation carriers: uncertainty and caution. J Natl Cancer Inst 100 (19): 1341-3, 2008. [PUBMED Abstract]
  140. Buys SS, Partridge E, Black A, et al.: Effect of screening on ovarian cancer mortality: the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Randomized Controlled Trial. JAMA 305 (22): 2295-303, 2011. [PUBMED Abstract]
  141. Hermsen BB, Olivier RI, Verheijen RH, et al.: No efficacy of annual gynaecological screening in BRCA1/2 mutation carriers; an observational follow-up study. Br J Cancer 96 (9): 1335-42, 2007. [PUBMED Abstract]
  142. Stirling D, Evans DG, Pichert G, et al.: Screening for familial ovarian cancer: failure of current protocols to detect ovarian cancer at an early stage according to the international Federation of gynecology and obstetrics system. J Clin Oncol 23 (24): 5588-96, 2005. [PUBMED Abstract]
  143. Olivier RI, Lubsen-Brandsma MA, Verhoef S, et al.: CA125 and transvaginal ultrasound monitoring in high-risk women cannot prevent the diagnosis of advanced ovarian cancer. Gynecol Oncol 100 (1): 20-6, 2006. [PUBMED Abstract]
  144. Meeuwissen PA, Seynaeve C, Brekelmans CT, et al.: Outcome of surveillance and prophylactic salpingo-oophorectomy in asymptomatic women at high risk for ovarian cancer. Gynecol Oncol 97 (2): 476-82, 2005. [PUBMED Abstract]
  145. Dørum A, Kristensen GB, Abeler VM, et al.: Early detection of familial ovarian cancer. Eur J Cancer 32A (10): 1645-51, 1996. [PUBMED Abstract]
  146. Tailor A, Bourne TH, Campbell S, et al.: Results from an ultrasound-based familial ovarian cancer screening clinic: a 10-year observational study. Ultrasound Obstet Gynecol 21 (4): 378-85, 2003. [PUBMED Abstract]
  147. Karlan BY, Raffel LJ, Crvenkovic G, et al.: A multidisciplinary approach to the early detection of ovarian carcinoma: rationale, protocol design, and early results. Am J Obstet Gynecol 169 (3): 494-501, 1993. [PUBMED Abstract]
  148. Muto MG, Cramer DW, Brown DL, et al.: Screening for ovarian cancer: the preliminary experience of a familial ovarian cancer center. Gynecol Oncol 51 (1): 12-20, 1993. [PUBMED Abstract]
  149. Liede A, Karlan BY, Baldwin RL, et al.: Cancer incidence in a population of Jewish women at risk of ovarian cancer. J Clin Oncol 20 (6): 1570-7, 2002. [PUBMED Abstract]
  150. Laframboise S, Nedelcu R, Murphy J, et al.: Use of CA-125 and ultrasound in high-risk women. Int J Gynecol Cancer 12 (1): 86-91, 2002 Jan-Feb. [PUBMED Abstract]
  151. Woodward ER, Sleightholme HV, Considine AM, et al.: Annual surveillance by CA125 and transvaginal ultrasound for ovarian cancer in both high-risk and population risk women is ineffective. BJOG 114 (12): 1500-9, 2007. [PUBMED Abstract]
  152. van der Velde NM, Mourits MJ, Arts HJ, et al.: Time to stop ovarian cancer screening in BRCA1/2 mutation carriers? Int J Cancer 124 (4): 919-23, 2009. [PUBMED Abstract]
  153. Rosenthal AN, Fraser L, Manchanda R, et al.: Results of annual screening in phase I of the United Kingdom familial ovarian cancer screening study highlight the need for strict adherence to screening schedule. J Clin Oncol 31 (1): 49-57, 2013. [PUBMED Abstract]
  154. Evans DG, Gaarenstroom KN, Stirling D, et al.: Screening for familial ovarian cancer: poor survival of BRCA1/2 related cancers. J Med Genet 46 (9): 593-7, 2009. [PUBMED Abstract]
  155. NIH consensus conference. Ovarian cancer. Screening, treatment, and follow-up. NIH Consensus Development Panel on Ovarian Cancer. JAMA 273 (6): 491-7, 1995. [PUBMED Abstract]
  156. Pepe MS, Etzioni R, Feng Z, et al.: Phases of biomarker development for early detection of cancer. J Natl Cancer Inst 93 (14): 1054-61, 2001. [PUBMED Abstract]
  157. Grosse SD, Khoury MJ: What is the clinical utility of genetic testing? Genet Med 8 (7): 448-50, 2006. [PUBMED Abstract]
  158. Finch A, Shaw P, Rosen B, et al.: Clinical and pathologic findings of prophylactic salpingo-oophorectomies in 159 BRCA1 and BRCA2 carriers. Gynecol Oncol 100 (1): 58-64, 2006. [PUBMED Abstract]
  159. Andersen MR, Goff BA, Lowe KA, et al.: Combining a symptoms index with CA 125 to improve detection of ovarian cancer. Cancer 113 (3): 484-9, 2008. [PUBMED Abstract]
  160. Skates SJ, Xu FJ, Yu YH, et al.: Toward an optimal algorithm for ovarian cancer screening with longitudinal tumor markers. Cancer 76 (10 Suppl): 2004-10, 1995. [PUBMED Abstract]
  161. Skates SJ, Menon U, MacDonald N, et al.: Calculation of the risk of ovarian cancer from serial CA-125 values for preclinical detection in postmenopausal women. J Clin Oncol 21 (10 Suppl): 206s-210s, 2003. [PUBMED Abstract]
  162. Menon U, Skates SJ, Lewis S, et al.: Prospective study using the risk of ovarian cancer algorithm to screen for ovarian cancer. J Clin Oncol 23 (31): 7919-26, 2005. [PUBMED Abstract]
  163. Greene MH, Piedmonte M, Alberts D, et al.: A prospective study of risk-reducing salpingo-oophorectomy and longitudinal CA-125 screening among women at increased genetic risk of ovarian cancer: design and baseline characteristics: a Gynecologic Oncology Group study. Cancer Epidemiol Biomarkers Prev 17 (3): 594-604, 2008. [PUBMED Abstract]
  164. Gagnon A, Ye B: Discovery and application of protein biomarkers for ovarian cancer. Curr Opin Obstet Gynecol 20 (1): 9-13, 2008. [PUBMED Abstract]
  165. Hennessy BT, Murph M, Nanjundan M, et al.: Ovarian cancer: linking genomics to new target discovery and molecular markers--the way ahead. Adv Exp Med Biol 617: 23-40, 2008. [PUBMED Abstract]
  166. Badgwell D, Bast RC: Early detection of ovarian cancer. Dis Markers 23 (5-6): 397-410, 2007. [PUBMED Abstract]
  167. Petricoin EF, Ardekani AM, Hitt BA, et al.: Use of proteomic patterns in serum to identify ovarian cancer. Lancet 359 (9306): 572-7, 2002. [PUBMED Abstract]
  168. Zhang Z, Bast RC, Yu Y, et al.: Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res 64 (16): 5882-90, 2004. [PUBMED Abstract]
  169. Koehn H, Oehler MK: Proteins' promise--progress and challenges in ovarian cancer proteomics. Menopause Int 13 (4): 148-53, 2007. [PUBMED Abstract]
  170. Visintin I, Feng Z, Longton G, et al.: Diagnostic markers for early detection of ovarian cancer. Clin Cancer Res 14 (4): 1065-72, 2008. [PUBMED Abstract]
  171. Simon R: Roadmap for developing and validating therapeutically relevant genomic classifiers. J Clin Oncol 23 (29): 7332-41, 2005. [PUBMED Abstract]
  172. Rutter JL, Wacholder S, Chetrit A, et al.: Gynecologic surgeries and risk of ovarian cancer in women with BRCA1 and BRCA2 Ashkenazi founder mutations: an Israeli population-based case-control study. J Natl Cancer Inst 95 (14): 1072-8, 2003. [PUBMED Abstract]
  173. Kotsopoulos J, Lubinski J, Lynch HT, et al.: Oophorectomy after menopause and the risk of breast cancer in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev 21 (7): 1089-96, 2012. [PUBMED Abstract]
  174. Sherman ME, Piedmonte M, Mai PL, et al.: Pathologic findings at risk-reducing salpingo-oophorectomy: primary results from Gynecologic Oncology Group Trial GOG-0199. J Clin Oncol 32 (29): 3275-83, 2014. [PUBMED Abstract]
  175. Domchek SM, Friebel TM, Neuhausen SL, et al.: Mortality after bilateral salpingo-oophorectomy in BRCA1 and BRCA2 mutation carriers: a prospective cohort study. Lancet Oncol 7 (3): 223-9, 2006. [PUBMED Abstract]
  176. Leeper K, Garcia R, Swisher E, et al.: Pathologic findings in prophylactic oophorectomy specimens in high-risk women. Gynecol Oncol 87 (1): 52-6, 2002. [PUBMED Abstract]
  177. Olivier RI, van Beurden M, Lubsen MA, et al.: Clinical outcome of prophylactic oophorectomy in BRCA1/BRCA2 mutation carriers and events during follow-up. Br J Cancer 90 (8): 1492-7, 2004. [PUBMED Abstract]
  178. Colgan TJ, Murphy J, Cole DE, et al.: Occult carcinoma in prophylactic oophorectomy specimens: prevalence and association with BRCA germline mutation status. Am J Surg Pathol 25 (10): 1283-9, 2001. [PUBMED Abstract]
  179. Powell CB, Kenley E, Chen LM, et al.: Risk-reducing salpingo-oophorectomy in BRCA mutation carriers: role of serial sectioning in the detection of occult malignancy. J Clin Oncol 23 (1): 127-32, 2005. [PUBMED Abstract]
  180. Callahan MJ, Crum CP, Medeiros F, et al.: Primary fallopian tube malignancies in BRCA-positive women undergoing surgery for ovarian cancer risk reduction. J Clin Oncol 25 (25): 3985-90, 2007. [PUBMED Abstract]
  181. Domchek SM, Friebel TM, Garber JE, et al.: Occult ovarian cancers identified at risk-reducing salpingo-oophorectomy in a prospective cohort of BRCA1/2 mutation carriers. Breast Cancer Res Treat 124 (1): 195-203, 2010. [PUBMED Abstract]
  182. Powell CB, Chen LM, McLennan J, et al.: Risk-reducing salpingo-oophorectomy (RRSO) in BRCA mutation carriers: experience with a consecutive series of 111 patients using a standardized surgical-pathological protocol. Int J Gynecol Cancer 21 (5): 846-51, 2011. [PUBMED Abstract]
  183. Piek JM, van Diest PJ, Zweemer RP, et al.: Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer. J Pathol 195 (4): 451-6, 2001. [PUBMED Abstract]
  184. Paley PJ, Swisher EM, Garcia RL, et al.: Occult cancer of the fallopian tube in BRCA-1 germline mutation carriers at prophylactic oophorectomy: a case for recommending hysterectomy at surgical prophylaxis. Gynecol Oncol 80 (2): 176-80, 2001. [PUBMED Abstract]
  185. Rose PG, Shrigley R, Wiesner GL: Germline BRCA2 mutation in a patient with fallopian tube carcinoma: a case report. Gynecol Oncol 77 (2): 319-20, 2000. [PUBMED Abstract]
  186. Zweemer RP, van Diest PJ, Verheijen RH, et al.: Molecular evidence linking primary cancer of the fallopian tube to BRCA1 germline mutations. Gynecol Oncol 76 (1): 45-50, 2000. [PUBMED Abstract]
  187. Piek JM, Torrenga B, Hermsen B, et al.: Histopathological characteristics of BRCA1- and BRCA2-associated intraperitoneal cancer: a clinic-based study. Fam Cancer 2 (2): 73-8, 2003. [PUBMED Abstract]
  188. Levine DA, Argenta PA, Yee CJ, et al.: Fallopian tube and primary peritoneal carcinomas associated with BRCA mutations. J Clin Oncol 21 (22): 4222-7, 2003. [PUBMED Abstract]
  189. Aziz S, Kuperstein G, Rosen B, et al.: A genetic epidemiological study of carcinoma of the fallopian tube. Gynecol Oncol 80 (3): 341-5, 2001. [PUBMED Abstract]
  190. Kindelberger DW, Lee Y, Miron A, et al.: Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: Evidence for a causal relationship. Am J Surg Pathol 31 (2): 161-9, 2007. [PUBMED Abstract]
  191. Rabban JT, Krasik E, Chen LM, et al.: Multistep level sections to detect occult fallopian tube carcinoma in risk-reducing salpingo-oophorectomies from women with BRCA mutations: implications for defining an optimal specimen dissection protocol. Am J Surg Pathol 33 (12): 1878-85, 2009. [PUBMED Abstract]
  192. Society of Gynecologic Oncologists Clinical Practice Committee Statement on Prophylactic Salpingo-oophorectomy. Gynecol Oncol 98 (2): 179-81, 2005. [PUBMED Abstract]
  193. Chen KT, Schooley JL, Flam MS: Peritoneal carcinomatosis after prophylactic oophorectomy in familial ovarian cancer syndrome. Obstet Gynecol 66 (3 Suppl): 93S-94S, 1985. [PUBMED Abstract]
  194. Lynch HT, Bewtra C, Lynch JF: Familial ovarian carcinoma. Clinical nuances. Am J Med 81 (6): 1073-6, 1986. [PUBMED Abstract]
  195. Lynch HT, Watson P, Bewtra C, et al.: Hereditary ovarian cancer. Heterogeneity in age at diagnosis. Cancer 67 (5): 1460-6, 1991. [PUBMED Abstract]
  196. Tobacman JK, Greene MH, Tucker MA, et al.: Intra-abdominal carcinomatosis after prophylactic oophorectomy in ovarian-cancer-prone families. Lancet 2 (8302): 795-7, 1982. [PUBMED Abstract]
  197. Truong LD, Maccato ML, Awalt H, et al.: Serous surface carcinoma of the peritoneum: a clinicopathologic study of 22 cases. Hum Pathol 21 (1): 99-110, 1990. [PUBMED Abstract]
  198. Piver MS, Jishi MF, Tsukada Y, et al.: Primary peritoneal carcinoma after prophylactic oophorectomy in women with a family history of ovarian cancer. A report of the Gilda Radner Familial Ovarian Cancer Registry. Cancer 71 (9): 2751-5, 1993. [PUBMED Abstract]
  199. 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 97 (2): 457-67, 2005. [PUBMED Abstract]
  200. 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 296 (2): 185-92, 2006. [PUBMED Abstract]
  201. Powell CB, Swisher EM, Cass I, et al.: Long term follow up of BRCA1 and BRCA2 mutation carriers with unsuspected neoplasia identified at risk reducing salpingo-oophorectomy. Gynecol Oncol 129 (2): 364-71, 2013. [PUBMED Abstract]
  202. Zakhour M, Danovitch Y, Lester J, et al.: Occult and subsequent cancer incidence following risk-reducing surgery in BRCA mutation carriers. Gynecol Oncol 143 (2): 231-235, 2016. [PUBMED Abstract]
  203. Chen S, Iversen ES, Friebel T, et al.: Characterization of BRCA1 and BRCA2 mutations in a large United States sample. J Clin Oncol 24 (6): 863-71, 2006. [PUBMED Abstract]
  204. 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 72 (5): 1117-30, 2003. [PUBMED Abstract]
  205. Risch HA, McLaughlin JR, Cole DE, et al.: Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario, Canada. J Natl Cancer Inst 98 (23): 1694-706, 2006. [PUBMED Abstract]
  206. Solsky I, Chen J, Rebbeck TR: Precision prophylaxis: Identifying the optimal timing for risk-reducing salpingo-oophorectomy based on type of BRCA1 and BRCA2 cluster region mutations. Gynecol Oncol 156 (2): 363-376, 2020. [PUBMED Abstract]
  207. Lavie O, Hornreich G, Ben-Arie A, et al.: BRCA germline mutations in Jewish women with uterine serous papillary carcinoma. Gynecol Oncol 92 (2): 521-4, 2004. [PUBMED Abstract]
  208. Goshen R, Chu W, Elit L, et al.: Is uterine papillary serous adenocarcinoma a manifestation of the hereditary breast-ovarian cancer syndrome? Gynecol Oncol 79 (3): 477-81, 2000. [PUBMED Abstract]
  209. Beiner ME, Finch A, Rosen B, et al.: The risk of endometrial cancer in women with BRCA1 and BRCA2 mutations. A prospective study. Gynecol Oncol 104 (1): 7-10, 2007. [PUBMED Abstract]
  210. 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 130 (1): 127-31, 2013. [PUBMED Abstract]
  211. Levine DA, Lin O, Barakat RR, et al.: Risk of endometrial carcinoma associated with BRCA mutation. Gynecol Oncol 80 (3): 395-8, 2001. [PUBMED Abstract]
  212. Karlan BY: Defining cancer risks for BRCA germline mutation carriers: implications for surgical prophylaxis. Gynecol Oncol 92 (2): 519-20, 2004. [PUBMED Abstract]
  213. 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 32 (10): 1097-100, 2006. [PUBMED Abstract]
  214. Lu KH, Kauff ND: Does a BRCA mutation plus tamoxifen equal hysterectomy? Gynecol Oncol 104 (1): 3-4, 2007. [PUBMED Abstract]
  215. Madalinska JB, Hollenstein J, Bleiker E, et al.: Quality-of-life effects of prophylactic salpingo-oophorectomy versus gynecologic screening among women at increased risk of hereditary ovarian cancer. J Clin Oncol 23 (28): 6890-8, 2005. [PUBMED Abstract]
  216. Rocca WA, Grossardt BR, de Andrade M, et al.: Survival patterns after oophorectomy in premenopausal women: a population-based cohort study. Lancet Oncol 7 (10): 821-8, 2006. [PUBMED Abstract]
  217. Rocca WA, Bower JH, Maraganore DM, et al.: Increased risk of parkinsonism in women who underwent oophorectomy before menopause. Neurology 70 (3): 200-9, 2008. [PUBMED Abstract]
  218. Shuster LT, Rhodes DJ, Gostout BS, et al.: Premature menopause or early menopause: long-term health consequences. Maturitas 65 (2): 161-6, 2010. [PUBMED Abstract]
  219. Parker WH, Broder MS, Chang E, et al.: Ovarian conservation at the time of hysterectomy and long-term health outcomes in the nurses' health study. Obstet Gynecol 113 (5): 1027-37, 2009. [PUBMED Abstract]
  220. Rivera CM, Grossardt BR, Rhodes DJ, et al.: Increased cardiovascular mortality after early bilateral oophorectomy. Menopause 16 (1): 15-23, 2009 Jan-Feb. [PUBMED Abstract]
  221. Michelsen TM, Pripp AH, Tonstad S, et al.: Metabolic syndrome after risk-reducing salpingo-oophorectomy in women at high risk for hereditary breast ovarian cancer: a controlled observational study. Eur J Cancer 45 (1): 82-9, 2009. [PUBMED Abstract]
  222. 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 204 (1): 19.e1-6, 2011. [PUBMED Abstract]
  223. Dietl J, Wischhusen J, Häusler SF: The post-reproductive Fallopian tube: better removed? Hum Reprod 26 (11): 2918-24, 2011. [PUBMED Abstract]
  224. Leblanc E, Narducci F, Farre I, et al.: Radical fimbriectomy: a reasonable temporary risk-reducing surgery for selected women with a germ line mutation of BRCA 1 or 2 genes? Rationale and preliminary development. Gynecol Oncol 121 (3): 472-6, 2011. [PUBMED Abstract]
  225. Kwon JS, Tinker A, Pansegrau G, et al.: Prophylactic salpingectomy and delayed oophorectomy as an alternative for BRCA mutation carriers. Obstet Gynecol 121 (1): 14-24, 2013. [PUBMED Abstract]
  226. Harmsen MG, IntHout J, Arts-de Jong M, et al.: Salpingectomy With Delayed Oophorectomy in BRCA1/2 Mutation Carriers: Estimating Ovarian Cancer Risk. Obstet Gynecol 127 (6): 1054-63, 2016. [PUBMED Abstract]
  227. Nebgen DR, Hurteau J, Holman LL, et al.: Bilateral salpingectomy with delayed oophorectomy for ovarian cancer risk reduction: A pilot study in women with BRCA1/2 mutations. Gynecol Oncol 150 (1): 79-84, 2018. [PUBMED Abstract]
  228. Collaborative Group on Epidemiological Studies of Ovarian Cancer, Beral V, Doll R, et al.: Ovarian cancer and oral contraceptives: collaborative reanalysis of data from 45 epidemiological studies including 23,257 women with ovarian cancer and 87,303 controls. Lancet 371 (9609): 303-14, 2008. [PUBMED Abstract]
  229. Narod SA, Sun P, Ghadirian P, et al.: Tubal ligation and risk of ovarian cancer in carriers of BRCA1 or BRCA2 mutations: a case-control study. Lancet 357 (9267): 1467-70, 2001. [PUBMED Abstract]
  230. Whittemore AS, Balise RR, Pharoah PD, et al.: Oral contraceptive use and ovarian cancer risk among carriers of BRCA1 or BRCA2 mutations. Br J Cancer 91 (11): 1911-5, 2004. [PUBMED Abstract]
  231. McGuire V, Felberg A, Mills M, et al.: Relation of contraceptive and reproductive history to ovarian cancer risk in carriers and noncarriers of BRCA1 gene mutations. Am J Epidemiol 160 (7): 613-8, 2004. [PUBMED Abstract]
  232. 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 8 (1): 26-34, 2007. [PUBMED Abstract]
  233. Modan B, Hartge P, Hirsh-Yechezkel G, et al.: Parity, oral contraceptives, and the risk of ovarian cancer among carriers and noncarriers of a BRCA1 or BRCA2 mutation. N Engl J Med 345 (4): 235-40, 2001. [PUBMED Abstract]
  234. Kotsopoulos J, Lubinski J, Gronwald J, et al.: Factors influencing ovulation and the risk of ovarian cancer in BRCA1 and BRCA2 mutation carriers. Int J Cancer 137 (5): 1136-46, 2015. [PUBMED Abstract]
  235. Risch HA: Hormonal etiology of epithelial ovarian cancer, with a hypothesis concerning the role of androgens and progesterone. J Natl Cancer Inst 90 (23): 1774-86, 1998. [PUBMED Abstract]
  236. Hankinson SE, Colditz GA, Hunter DJ, et al.: A prospective study of reproductive factors and risk of epithelial ovarian cancer. Cancer 76 (2): 284-90, 1995. [PUBMED Abstract]
  237. Gronwald J, Byrski T, Huzarski T, et al.: Influence of selected lifestyle factors on breast and ovarian cancer risk in BRCA1 mutation carriers from Poland. Breast Cancer Res Treat 95 (2): 105-9, 2006. [PUBMED Abstract]
  238. Whittemore AS, Harris R, Itnyre J: Characteristics relating to ovarian cancer risk: collaborative analysis of 12 US case-control studies. IV. The pathogenesis of epithelial ovarian cancer. Collaborative Ovarian Cancer Group. Am J Epidemiol 136 (10): 1212-20, 1992. [PUBMED Abstract]
  239. Miracle-McMahill HL, Calle EE, Kosinski AS, et al.: Tubal ligation and fatal ovarian cancer in a large prospective cohort study. Am J Epidemiol 145 (4): 349-57, 1997. [PUBMED Abstract]
  240. Cancer risks in BRCA2 mutation carriers. The Breast Cancer Linkage Consortium. J Natl Cancer Inst 91 (15): 1310-6, 1999. [PUBMED Abstract]
  241. van Asperen CJ, Brohet RM, Meijers-Heijboer EJ, et al.: Cancer risks in BRCA2 families: estimates for sites other than breast and ovary. J Med Genet 42 (9): 711-9, 2005. [PUBMED Abstract]
  242. Risch HA, McLaughlin JR, Cole DE, et al.: Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a population series of 649 women with ovarian cancer. Am J Hum Genet 68 (3): 700-10, 2001. [PUBMED Abstract]
  243. Liede A, Malik IA, Aziz Z, et al.: Contribution of BRCA1 and BRCA2 mutations to breast and ovarian cancer in Pakistan. Am J Hum Genet 71 (3): 595-606, 2002. [PUBMED Abstract]
  244. 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 66 (4): 1259-72, 2000. [PUBMED Abstract]
  245. Mohamad HB, Apffelstaedt JP: Counseling for male BRCA mutation carriers: a review. Breast 17 (5): 441-50, 2008. [PUBMED Abstract]
  246. Liede A, Karlan BY, Narod SA: Cancer risks for male carriers of germline mutations in BRCA1 or BRCA2: a review of the literature. J Clin Oncol 22 (4): 735-42, 2004. [PUBMED Abstract]
  247. 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 98 (2): 502-7, 2008. [PUBMED Abstract]
  248. Tryggvadóttir L, Vidarsdóttir L, Thorgeirsson T, et al.: Prostate cancer progression and survival in BRCA2 mutation carriers. J Natl Cancer Inst 99 (12): 929-35, 2007. [PUBMED Abstract]
  249. Agalliu I, Gern R, Leanza S, et al.: Associations of high-grade prostate cancer with BRCA1 and BRCA2 founder mutations. Clin Cancer Res 15 (3): 1112-20, 2009. [PUBMED Abstract]
  250. Narod SA, Neuhausen S, Vichodez G, et al.: Rapid progression of prostate cancer in men with a BRCA2 mutation. Br J Cancer 99 (2): 371-4, 2008. [PUBMED Abstract]
  251. Edwards SM, Evans DG, Hope Q, et al.: Prostate cancer in BRCA2 germline mutation carriers is associated with poorer prognosis. Br J Cancer 103 (6): 918-24, 2010. [PUBMED Abstract]
  252. Gallagher DJ, Gaudet MM, Pal P, et al.: Germline BRCA mutations denote a clinicopathologic subset of prostate cancer. Clin Cancer Res 16 (7): 2115-21, 2010. [PUBMED Abstract]
  253. Schröder FH, Hugosson J, Roobol MJ, et al.: Screening and prostate-cancer mortality in a randomized European study. N Engl J Med 360 (13): 1320-8, 2009. [PUBMED Abstract]
  254. Andriole GL, Grubb RL, Buys SS, et al.: Mortality results from a randomized prostate-cancer screening trial. N Engl J Med 360 (13): 1310-9, 2009. [PUBMED Abstract]
  255. Fiorentino M, Judson G, Penney K, et al.: Immunohistochemical expression of BRCA1 and lethal prostate cancer. Cancer Res 70 (8): 3136-9, 2010. [PUBMED Abstract]
  256. Horsburgh S, Matthew A, Bristow R, et al.: Male BRCA1 and BRCA2 mutation carriers: a pilot study investigating medical characteristics of patients participating in a prostate cancer prevention clinic. Prostate 65 (2): 124-9, 2005. [PUBMED Abstract]
  257. Hubert A, Peretz T, Manor O, et al.: The Jewish Ashkenazi founder mutations in the BRCA1/BRCA2 genes are not found at an increased frequency in Ashkenazi patients with prostate cancer. Am J Hum Genet 65 (3): 921-4, 1999. [PUBMED Abstract]
  258. 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 107 (1): 28-39, 2011. [PUBMED Abstract]
  259. Foulkes WD, Metcalfe K, Hanna W, et al.: Disruption of the expected positive correlation between breast tumor size and lymph node status in BRCA1-related breast carcinoma. Cancer 98 (8): 1569-77, 2003. [PUBMED Abstract]
  260. Verhoog LC, Brekelmans CT, Seynaeve C, et al.: Survival and tumour characteristics of breast-cancer patients with germline mutations of BRCA1. Lancet 351 (9099): 316-21, 1998. [PUBMED Abstract]
  261. Jóhannsson OT, Ranstam J, Borg A, et al.: Survival of BRCA1 breast and ovarian cancer patients: a population-based study from southern Sweden. J Clin Oncol 16 (2): 397-404, 1998. [PUBMED Abstract]
  262. Stoppa-Lyonnet D, Ansquer Y, Dreyfus H, et al.: Familial invasive breast cancers: worse outcome related to BRCA1 mutations. J Clin Oncol 18 (24): 4053-9, 2000. [PUBMED Abstract]
  263. Haffty BG, Harrold E, Khan AJ, et al.: Outcome of conservatively managed early-onset breast cancer by BRCA1/2 status. Lancet 359 (9316): 1471-7, 2002. [PUBMED Abstract]
  264. Robson M, Levin D, Federici M, et al.: Breast conservation therapy for invasive breast cancer in Ashkenazi women with BRCA gene founder mutations. J Natl Cancer Inst 91 (24): 2112-7, 1999. [PUBMED Abstract]
  265. Graeser MK, Engel C, Rhiem K, et al.: Contralateral breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Clin Oncol 27 (35): 5887-92, 2009. [PUBMED Abstract]
  266. van den Broek AJ, van 't Veer LJ, Hooning MJ, et al.: Impact of Age at Primary Breast Cancer on Contralateral Breast Cancer Risk in BRCA1/2 Mutation Carriers. J Clin Oncol 34 (5): 409-18, 2016. [PUBMED Abstract]
  267. Menes TS, Terry MB, Goldgar D, et al.: Second primary breast cancer in BRCA1 and BRCA2 mutation carriers: 10-year cumulative incidence in the Breast Cancer Family Registry. Breast Cancer Res Treat 151 (3): 653-60, 2015. [PUBMED Abstract]
  268. Robson ME, Chappuis PO, Satagopan J, et al.: A combined analysis of outcome following breast cancer: differences in survival based on BRCA1/BRCA2 mutation status and administration of adjuvant treatment. Breast Cancer Res 6 (1): R8-R17, 2004. [PUBMED Abstract]
  269. Rennert G, Bisland-Naggan S, Barnett-Griness O, et al.: Clinical outcomes of breast cancer in carriers of BRCA1 and BRCA2 mutations. N Engl J Med 357 (2): 115-23, 2007. [PUBMED Abstract]
  270. Kriege M, Seynaeve C, Meijers-Heijboer H, et al.: Sensitivity to first-line chemotherapy for metastatic breast cancer in BRCA1 and BRCA2 mutation carriers. J Clin Oncol 27 (23): 3764-71, 2009. [PUBMED Abstract]
  271. Goodwin PJ, Phillips KA, West DW, et al.: Breast cancer prognosis in BRCA1 and BRCA2 mutation carriers: an International Prospective Breast Cancer Family Registry population-based cohort study. J Clin Oncol 30 (1): 19-26, 2012. [PUBMED Abstract]
  272. 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 19 (2): 169-180, 2018. [PUBMED Abstract]
  273. Gonzalez-Angulo AM, Timms KM, Liu S, et al.: Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 17 (5): 1082-9, 2011. [PUBMED Abstract]
  274. Lee LJ, Alexander B, Schnitt SJ, et al.: Clinical outcome of triple negative breast cancer in BRCA1 mutation carriers and noncarriers. Cancer 117 (14): 3093-100, 2011. [PUBMED Abstract]
  275. Tung N, Gaughan E, Hacker MR, et al.: Outcome of triple negative breast cancer: comparison of sporadic and BRCA1-associated cancers. Breast Cancer Res Treat 146 (1): 175-82, 2014. [PUBMED Abstract]
  276. Huzarski T, Byrski T, Gronwald J, et al.: Ten-year survival in patients with BRCA1-negative and BRCA1-positive breast cancer. J Clin Oncol 31 (26): 3191-6, 2013. [PUBMED Abstract]
  277. Verhoog LC, Berns EM, Brekelmans CT, et al.: Prognostic significance of germline BRCA2 mutations in hereditary breast cancer patients. J Clin Oncol 18 (21 Suppl): 119S-24S, 2000. [PUBMED Abstract]
  278. Brekelmans CT, Tilanus-Linthorst MM, Seynaeve C, et al.: Tumour characteristics, survival and prognostic factors of hereditary breast cancer from BRCA2-, BRCA1- and non-BRCA1/2 families as compared to sporadic breast cancer cases. Eur J Cancer 43 (5): 867-76, 2007. [PUBMED Abstract]
  279. Budroni M, Cesaraccio R, Coviello V, et al.: Role of BRCA2 mutation status on overall survival among breast cancer patients from Sardinia. BMC Cancer 9: 62, 2009. [PUBMED Abstract]
  280. Byrski T, Huzarski T, Dent R, et al.: Response to neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat 115 (2): 359-63, 2009. [PUBMED Abstract]
  281. Chappuis PO, Goffin J, Wong N, et al.: A significant response to neoadjuvant chemotherapy in BRCA1/2 related breast cancer. J Med Genet 39 (8): 608-10, 2002. [PUBMED Abstract]
  282. 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 28 (3): 375-9, 2010. [PUBMED Abstract]
  283. Fourquet A, Stoppa-Lyonnet D, Kirova YM, et al.: Familial breast cancer: clinical response to induction chemotherapy or radiotherapy related to BRCA1/2 mutations status. Am J Clin Oncol 32 (2): 127-31, 2009. [PUBMED Abstract]
  284. Arun B, Bayraktar S, Liu DD, et al.: Response to neoadjuvant systemic therapy for breast cancer in BRCA mutation carriers and noncarriers: a single-institution experience. J Clin Oncol 29 (28): 3739-46, 2011. [PUBMED Abstract]
  285. Wunderle M, Gass P, Häberle L, et al.: BRCA mutations and their influence on pathological complete response and prognosis in a clinical cohort of neoadjuvantly treated breast cancer patients. Breast Cancer Res Treat 171 (1): 85-94, 2018. [PUBMED Abstract]
  286. Silver DP, Richardson AL, Eklund AC, et al.: Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol 28 (7): 1145-53, 2010. [PUBMED Abstract]
  287. Byrski T, Huzarski T, Dent R, et al.: Pathologic complete response to neoadjuvant cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res Treat 147 (2): 401-5, 2014. [PUBMED Abstract]
  288. Hahnen E, Lederer B, Hauke J, et al.: Germline Mutation Status, Pathological Complete Response, and Disease-Free Survival in Triple-Negative Breast Cancer: Secondary Analysis of the GeparSixto Randomized Clinical Trial. JAMA Oncol 3 (10): 1378-1385, 2017. [PUBMED Abstract]
  289. Tutt A, Tovey H, Cheang MCU, et al.: Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat Med 24 (5): 628-637, 2018. [PUBMED Abstract]
  290. Robson M, Im SA, Senkus E, et al.: Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. N Engl J Med 377 (6): 523-533, 2017. [PUBMED Abstract]
  291. Litton JK, Rugo HS, Ettl J, et al.: Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N Engl J Med 379 (8): 753-763, 2018. [PUBMED Abstract]
  292. Tutt ANJ, Garber JE, Kaufman B, et al.: Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N Engl J Med 384 (25): 2394-2405, 2021. [PUBMED Abstract]
  293. Leong T, Whitty J, Keilar M, et al.: Mutation analysis of BRCA1 and BRCA2 cancer predisposition genes in radiation hypersensitive cancer patients. Int J Radiat Oncol Biol Phys 48 (4): 959-65, 2000. [PUBMED Abstract]
  294. Pierce LJ, Strawderman M, Narod SA, et al.: Effect of radiotherapy after breast-conserving treatment in women with breast cancer and germline BRCA1/2 mutations. J Clin Oncol 18 (19): 3360-9, 2000. [PUBMED Abstract]
  295. Shanley S, McReynolds K, Ardern-Jones A, et al.: Late toxicity is not increased in BRCA1/BRCA2 mutation carriers undergoing breast radiotherapy in the United Kingdom. Clin Cancer Res 12 (23): 7025-32, 2006. [PUBMED Abstract]
  296. Pierce LJ, Phillips KA, Griffith KA, et al.: Local therapy in BRCA1 and BRCA2 mutation carriers with operable breast cancer: comparison of breast conservation and mastectomy. Breast Cancer Res Treat 121 (2): 389-98, 2010. [PUBMED Abstract]
  297. Kirova YM, Savignoni A, Sigal-Zafrani B, et al.: Is the breast-conserving treatment with radiotherapy appropriate in BRCA1/2 mutation carriers? Long-term results and review of the literature. Breast Cancer Res Treat 120 (1): 119-26, 2010. [PUBMED Abstract]
  298. Metcalfe K, Lynch HT, Ghadirian P, et al.: Risk of ipsilateral breast cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat 127 (1): 287-96, 2011. [PUBMED Abstract]
  299. Cao W, Xie Y, He Y, et al.: Risk of ipsilateral breast tumor recurrence in primary invasive breast cancer following breast-conserving surgery with BRCA1 and BRCA2 mutation in China. Breast Cancer Res Treat 175 (3): 749-754, 2019. [PUBMED Abstract]
  300. Bernstein JL, Thomas DC, Shore RE, et al.: Contralateral breast cancer after radiotherapy among BRCA1 and BRCA2 mutation carriers: a WECARE study report. Eur J Cancer 49 (14): 2979-85, 2013. [PUBMED Abstract]
  301. Rubin SC, Benjamin I, Behbakht K, et al.: Clinical and pathological features of ovarian cancer in women with germ-line mutations of BRCA1. N Engl J Med 335 (19): 1413-6, 1996. [PUBMED Abstract]
  302. Ben David Y, Chetrit A, Hirsh-Yechezkel G, et al.: Effect of BRCA mutations on the length of survival in epithelial ovarian tumors. J Clin Oncol 20 (2): 463-6, 2002. [PUBMED Abstract]
  303. Jazaeri AA, Yee CJ, Sotiriou C, et al.: Gene expression profiles of BRCA1-linked, BRCA2-linked, and sporadic ovarian cancers. J Natl Cancer Inst 94 (13): 990-1000, 2002. [PUBMED Abstract]
  304. Cass I, Baldwin RL, Varkey T, et al.: Improved survival in women with BRCA-associated ovarian carcinoma. Cancer 97 (9): 2187-95, 2003. [PUBMED Abstract]
  305. Aida H, Takakuwa K, Nagata H, et al.: Clinical features of ovarian cancer in Japanese women with germ-line mutations of BRCA1. Clin Cancer Res 4 (1): 235-40, 1998. [PUBMED Abstract]
  306. Boyd J, Sonoda Y, Federici MG, et al.: Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. JAMA 283 (17): 2260-5, 2000. [PUBMED Abstract]
  307. Tan DS, Rothermundt C, Thomas K, et al.: "BRCAness" syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol 26 (34): 5530-6, 2008. [PUBMED Abstract]
  308. Hyman DM, Zhou Q, Iasonos A, et al.: Improved survival for BRCA2-associated serous ovarian cancer compared with both BRCA-negative and BRCA1-associated serous ovarian cancer. Cancer 118 (15): 3703-9, 2012. [PUBMED Abstract]
  309. Liu J, Cristea MC, Frankel P, et al.: Clinical characteristics and outcomes of BRCA-associated ovarian cancer: genotype and survival. Cancer Genet 205 (1-2): 34-41, 2012 Jan-Feb. [PUBMED Abstract]
  310. Chetrit A, Hirsh-Yechezkel G, Ben-David Y, et al.: Effect of BRCA1/2 mutations on long-term survival of patients with invasive ovarian cancer: the national Israeli study of ovarian cancer. J Clin Oncol 26 (1): 20-5, 2008. [PUBMED Abstract]
  311. Lacour RA, Westin SN, Meyer LA, et al.: Improved survival in non-Ashkenazi Jewish ovarian cancer patients with BRCA1 and BRCA2 gene mutations. Gynecol Oncol 121 (2): 358-63, 2011. [PUBMED Abstract]
  312. Vencken PM, Kriege M, Hoogwerf D, et al.: Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol 22 (6): 1346-52, 2011. [PUBMED Abstract]
  313. Pal T, Permuth-Wey J, Kapoor R, et al.: Improved survival in BRCA2 carriers with ovarian cancer. Fam Cancer 6 (1): 113-9, 2007. [PUBMED Abstract]
  314. Yang D, Khan S, Sun Y, et al.: Association of BRCA1 and BRCA2 mutations with survival, chemotherapy sensitivity, and gene mutator phenotype in patients with ovarian cancer. JAMA 306 (14): 1557-65, 2011. [PUBMED Abstract]
  315. Bolton KL, Chenevix-Trench G, Goh C, et al.: Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA 307 (4): 382-90, 2012. [PUBMED Abstract]
  316. Zweemer RP, Verheijen RH, Coebergh JW, et al.: Survival analysis in familial ovarian cancer, a case control study. Eur J Obstet Gynecol Reprod Biol 98 (2): 219-23, 2001. [PUBMED Abstract]
  317. Pharoah PD, Easton DF, Stockton DL, et al.: Survival in familial, BRCA1-associated, and BRCA2-associated epithelial ovarian cancer. United Kingdom Coordinating Committee for Cancer Research (UKCCCR) Familial Ovarian Cancer Study Group. Cancer Res 59 (4): 868-71, 1999. [PUBMED Abstract]
  318. Buller RE, Shahin MS, Geisler JP, et al.: Failure of BRCA1 dysfunction to alter ovarian cancer survival. Clin Cancer Res 8 (5): 1196-202, 2002. [PUBMED Abstract]
  319. McLaughlin JR, Rosen B, Moody J, et al.: Long-term ovarian cancer survival associated with mutation in BRCA1 or BRCA2. J Natl Cancer Inst 105 (2): 141-8, 2013. [PUBMED Abstract]
  320. Yang H, Jeffrey PD, Miller J, et al.: BRCA2 function in DNA binding and recombination from a BRCA2-DSS1-ssDNA structure. Science 297 (5588): 1837-48, 2002. [PUBMED Abstract]
  321. Xu X, Weaver Z, Linke SP, et al.: Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells. Mol Cell 3 (3): 389-95, 1999. [PUBMED Abstract]
  322. Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature 474 (7353): 609-15, 2011. [PUBMED Abstract]
  323. Husain A, He G, Venkatraman ES, et al.: BRCA1 up-regulation is associated with repair-mediated resistance to cis-diamminedichloroplatinum(II). Cancer Res 58 (6): 1120-3, 1998. [PUBMED Abstract]
  324. Pennington KP, Walsh T, Harrell MI, et al.: Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin Cancer Res 20 (3): 764-75, 2014. [PUBMED Abstract]
  325. Norquist BM, Brady MF, Harrell MI, et al.: Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study. Clin Cancer Res 24 (4): 777-783, 2018. [PUBMED Abstract]
  326. Fong PC, Boss DS, Yap TA, et al.: Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361 (2): 123-34, 2009. [PUBMED Abstract]
  327. Audeh MW, Carmichael J, Penson RT, et al.: Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet 376 (9737): 245-51, 2010. [PUBMED Abstract]
  328. Fong PC, Yap TA, Boss DS, et al.: Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol 28 (15): 2512-9, 2010. [PUBMED Abstract]
  329. Hennessy BT, Timms KM, Carey MS, et al.: Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol 28 (22): 3570-6, 2010. [PUBMED Abstract]
  330. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al.: Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 33 (3): 244-50, 2015. [PUBMED Abstract]
  331. Domchek SM, Aghajanian C, Shapira-Frommer R, et al.: Efficacy and safety of olaparib monotherapy in germline BRCA1/2 mutation carriers with advanced ovarian cancer and three or more lines of prior therapy. Gynecol Oncol 140 (2): 199-203, 2016. [PUBMED Abstract]
  332. Oza AM, Cibula D, Benzaquen AO, et al.: Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: a randomised phase 2 trial. Lancet Oncol 16 (1): 87-97, 2015. [PUBMED Abstract]
  333. Liu JF, Barry WT, Birrer M, et al.: Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum-sensitive ovarian cancer: a randomised phase 2 study. Lancet Oncol 15 (11): 1207-14, 2014. [PUBMED Abstract]
  334. Kaye SB, Lubinski J, Matulonis U, et al.: Phase II, open-label, randomized, multicenter study comparing the efficacy and safety of olaparib, a poly (ADP-ribose) polymerase inhibitor, and pegylated liposomal doxorubicin in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer. J Clin Oncol 30 (4): 372-9, 2012. [PUBMED Abstract]
  335. Adams SF, Marsh EB, Elmasri W, et al.: A high response rate to liposomal doxorubicin is seen among women with BRCA mutations treated for recurrent epithelial ovarian cancer. Gynecol Oncol 123 (3): 486-91, 2011. [PUBMED Abstract]
  336. Safra T, Borgato L, Nicoletto MO, et al.: BRCA mutation status and determinant of outcome in women with recurrent epithelial ovarian cancer treated with pegylated liposomal doxorubicin. Mol Cancer Ther 10 (10): 2000-7, 2011. [PUBMED Abstract]
  337. Gelmon KA, Tischkowitz M, Mackay H, et al.: Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study. Lancet Oncol 12 (9): 852-61, 2011. [PUBMED Abstract]
  338. Ledermann J, Harter P, Gourley C, et al.: Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med 366 (15): 1382-92, 2012. [PUBMED Abstract]
  339. Ledermann J, Harter P, Gourley C, et al.: Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 15 (8): 852-61, 2014. [PUBMED Abstract]
  340. Matulonis UA, Harter P, Gourley C, et al.: Olaparib maintenance therapy in patients with platinum-sensitive, relapsed serous ovarian cancer and a BRCA mutation: Overall survival adjusted for postprogression poly(adenosine diphosphate ribose) polymerase inhibitor therapy. Cancer 122 (12): 1844-52, 2016. [PUBMED Abstract]
  341. Ledermann JA, Harter P, Gourley C, et al.: Overall survival in patients with platinum-sensitive recurrent serous ovarian cancer receiving olaparib maintenance monotherapy: an updated analysis from a randomised, placebo-controlled, double-blind, phase 2 trial. Lancet Oncol 17 (11): 1579-1589, 2016. [PUBMED Abstract]
  342. Pujade-Lauraine E, Ledermann JA, Selle F, et al.: Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 18 (9): 1274-1284, 2017. [PUBMED Abstract]
  343. Moore K, Colombo N, Scambia G, et al.: Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 379 (26): 2495-2505, 2018. [PUBMED Abstract]
  344. Drew Y, Ledermann J, Hall G, et al.: Phase 2 multicentre trial investigating intermittent and continuous dosing schedules of the poly(ADP-ribose) polymerase inhibitor rucaparib in germline BRCA mutation carriers with advanced ovarian and breast cancer. Br J Cancer 114 (7): 723-30, 2016. [PUBMED Abstract]
  345. Kristeleit R, Shapiro GI, Burris HA, et al.: A Phase I-II Study of the Oral PARP Inhibitor Rucaparib in Patients with Germline BRCA1/2-Mutated Ovarian Carcinoma or Other Solid Tumors. Clin Cancer Res 23 (15): 4095-4106, 2017. [PUBMED Abstract]
  346. Swisher EM, Lin KK, Oza AM, et al.: Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol 18 (1): 75-87, 2017. [PUBMED Abstract]
  347. Coleman RL, Oza AM, Lorusso D, et al.: Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 390 (10106): 1949-1961, 2017. [PUBMED Abstract]
  348. Sandhu SK, Schelman WR, Wilding G, et al.: The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol 14 (9): 882-92, 2013. [PUBMED Abstract]
  349. Mirza MR, Monk BJ, Herrstedt J, et al.: Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med 375 (22): 2154-2164, 2016. [PUBMED Abstract]
  350. Moore KN, Secord AA, Geller MA, et al.: Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol 20 (5): 636-648, 2019. [PUBMED Abstract]
  351. Hampel H, Bennett RL, Buchanan A, et al.: A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med 17 (1): 70-87, 2015. [PUBMED Abstract]
  352. Committee on Practice Bulletins–Gynecology, Committee on Genetics, Society of Gynecologic Oncology: Practice Bulletin No 182: Hereditary Breast and Ovarian Cancer Syndrome. Obstet Gynecol 130 (3): e110-e126, 2017. [PUBMED Abstract]
  353. Robson ME, Bradbury AR, Arun B, et al.: American Society of Clinical Oncology Policy Statement Update: Genetic and Genomic Testing for Cancer Susceptibility. J Clin Oncol 33 (31): 3660-7, 2015. [PUBMED Abstract]
  354. Paluch-Shimon S, Cardoso F, Sessa C, et al.: Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO Clinical Practice Guidelines for cancer prevention and screening. Ann Oncol 27 (suppl 5): v103-v110, 2016. [PUBMED Abstract]
  355. American College of Surgeons: National Accreditation Program for Breast Centers Standards Manual 2014 Edition. Chicago, IL: American College of Surgeons, 2014. Available online. Last accessed February 4, 2022.
  356. Berliner JL, Fay AM, Cummings SA, et al.: NSGC practice guideline: risk assessment and genetic counseling for hereditary breast and ovarian cancer. J Genet Couns 22 (2): 155-63, 2013. [PUBMED Abstract]
  357. Lancaster JM, Powell CB, Chen LM, et al.: Society of Gynecologic Oncology statement on risk assessment for inherited gynecologic cancer predispositions. Gynecol Oncol 136 (1): 3-7, 2015. [PUBMED Abstract]
  358. Owens DK, Davidson KW, Krist AH, et al.: Risk Assessment, Genetic Counseling, and Genetic Testing for BRCA-Related Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 322 (7): 652-665, 2019. [PUBMED Abstract]

Clinical Management of Other Hereditary Breast and/or Gynecologic Cancer Syndromes

Lynch Syndrome

As mismatch repair genes were identified as the genetic basis of Lynch syndrome, microsatellite instability was identified as a common molecular marker of mismatch repair deficiency. Approximately 15% of sporadic colorectal cancers show microsatellite instability, while up to 28% of sporadic endometrial cancers have this molecular change.[1,2] Most frequently, sporadic tumors with microsatellite instability have hypermethylation of the MLH1 promoter. In Lynch syndrome–related tumors showing microsatellite instability, there is typically loss of one or more of the proteins associated with the mismatch repair genes.

Certain histopathologic features are also strongly suggestive of a microsatellite instability phenotype, including the presence of tumor infiltrating lymphocytes, peritumoral lymphocytes, undifferentiated carcinomas, and lower uterine segment tumors. Use of clinical criteria is one strategy of selection criteria for tumor testing. Computer models have also been used to predict the probability of a mismatch repair genetic variant and can be used in the absence of microsatellite instability or immunohistochemistry information.[3-6] Overall, however, there is a move towards universal testing of colorectal and endometrial tumors when tumor tissue is available. (Refer to the Universal tumor testing to screen for Lynch syndrome section in the PDQ summary on Genetics of Colorectal Cancer for more information.)

References
  1. Vilar E, Gruber SB: Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol 7 (3): 153-62, 2010. [PUBMED Abstract]
  2. Nakamura A, Osonoi T, Terauchi Y: Relationship between urinary sodium excretion and pioglitazone-induced edema. J Diabetes Investig 1 (5): 208-11, 2010. [PUBMED Abstract]
  3. Balmaña J, Stockwell DH, Steyerberg EW, et al.: Prediction of MLH1 and MSH2 mutations in Lynch syndrome. JAMA 296 (12): 1469-78, 2006. [PUBMED Abstract]
  4. Barnetson RA, Tenesa A, Farrington SM, et al.: Identification and survival of carriers of mutations in DNA mismatch-repair genes in colon cancer. N Engl J Med 354 (26): 2751-63, 2006. [PUBMED Abstract]
  5. Kastrinos F, Allen JI, Stockwell DH, et al.: Development and validation of a colon cancer risk assessment tool for patients undergoing colonoscopy. Am J Gastroenterol 104 (6): 1508-18, 2009. [PUBMED Abstract]
  6. Khan O, Blanco A, Conrad P, et al.: Performance of Lynch syndrome predictive models in a multi-center US referral population. Am J Gastroenterol 106 (10): 1822-7; quiz 1828, 2011. [PUBMED Abstract]

Psychosocial Issues in BRCA1/BRCA2

Introduction

Psychosocial research in the context of cancer genetic testing helps to define psychological outcomes, interpersonal and familial effects, and cultural and community responses. This type of research also identifies behavioral factors that encourage or impede screening and other health behaviors. It can enhance decision making about risk-reduction interventions, evaluate psychosocial interventions to reduce distress and/or other negative sequelae related to risk notification and genetic testing, provide data to help resolve ethical concerns, and predict the interest in testing of various groups.

This section addresses psychosocial issues in BRCA1/BRCA2. Psychosocial and screening issues related to gynecologic cancers associated with Lynch syndrome are discussed in the Psychosocial Issues in Hereditary Colon Cancer Syndromes section in the PDQ summary on Genetics of Colorectal Cancer.

Uptake of Genetic Counseling and Genetic Testing

Degree of uptake of genetic counseling and genetic testing

Comparison of uptake rates among studies in which counseling and testing were offered is challenging because of differences in methodologies, including the sampling strategy used, the recruitment setting, and testing through a research protocol with high-risk cohorts or kindreds. In a systematic review of 40 studies conducted before 2002 that had assessed genetic testing utilization, uptake rates varied widely and ranged from 25% to 96%, with an average uptake rate of 59%.[1] Results of multivariate analysis found that BRCA1/BRCA2 genetic testing uptake was associated with having a personal or family history of breast or ovarian cancer, and with methodological features of the studies, including sampling strategies, recruitment settings, and how studies defined actual uptake versus the intention to have testing.

Other factors have been positively correlated with uptake of BRCA1/BRCA2 genetic testing, although these findings are not consistent across all studies. Psychological factors that have been positively correlated with testing uptake include greater cancer-specific distress and greater perceived risk of developing breast or ovarian cancer. Having more cancer-affected relatives also has been correlated with greater testing uptake.

Table 14 summarizes the uptake of genetic testing in clinical and research cohorts in the United States.

Table 14. Predictors Associated with Uptake of Genetic Testing (GT)
Study Citation Study Population Sample Size (N) Uptake of GT Predictors Associated With Uptake of GT Comments
GC = genetic counseling; HMO = health maintenance organization.
aSelf-report as data source.
bMedical records as data source.
Schwartz et al. (2005) [2] Newly diagnosed and locally untreated breast cancer patients with ≥10% risk of having a BRCA1/BRCA2 pathogenic variant a 231 177/231 (77%) underwent GT Having decided on definitive local treatment. Women who were undecided on a definitive local treatment were more likely to be tested Testing was offered free of charge
34/231 (15%) had baseline interview but declined GT
Physician recommendation for testing. Women whose physician had recommended GT were more likely to be tested 38/177 chose to proceed with treatment before receiving test results
20/231 declined baseline interview
Kieran et al. (2007) [3] Women who received GC between 2002 and 2004a 250 88/250 (35%) underwent GT Ability to pay for GT (entire cost or cost not covered by insurance). Nonuptake was 5.5 times more likely in women who could not afford testing 450 women received GC for breast and ovarian cancer risk during study period. 250 women were retrospectively identified as eligible and were mailed a study questionnaire
36/88 returned surveys
Ability to recall risk estimates that were provided post-GC. Nonuptake was 15.5 times more likely in women who could not recall their risk estimates All women had some form of insurance
162/250 (65%) eligible
65/162 returned surveys
Susswein et al. (2008) [4] African American women and White women with breast cancerb 768 529/768 (69%) underwent GT Race and ethnicity. African American women were less likely to be tested than White women Sample obtained from a clinical database. Testing was offered free of charge when it was not covered by insurance. This effect for time of diagnosis was significant in the African American subgroup but not in the White subgroup
African American women: 77/132 (58%) underwent GT
Recent diagnosis. African American women who were recently diagnosed were more likely to be tested
White women: 452/636 (71%) underwent GT
Olaya et al. (2009) [5] Patients referred for GT between 2001 and 2008b 213 111/213 (52%) underwent GT Personal history of breast cancer. Having a personal history was associated with 3 times greater odds of being tested Insurance coverage for testing was available for 91.1% (175/213) of patients. Of those who had coverage for GT, 51.4% underwent testing and 48.6% did not. Of those without coverage, 41.2% had GT and 58.9% did not
102/213 (48%) declined GT Higher level of education. Those with a high school education or less had one-third the odds of being tested, compared with those with at least some college
Levy et al. (2010) [6] Women aged 20–40 y with newly diagnosed early-onset breast cancer.b 1,474 446/1,474 (30%) underwent GT Race and ethnicity. Women of Jewish ethnicity were 3 times more likely to be tested than non-Jewish White women. African American and Hispanic women were significantly less likely to receive testing than non-Jewish White women Sample obtained from a national database of commercially insured individuals
Jewish women: 18/32 (56%) underwent GT Home location. Women living in the south were more likely to be tested than women living in the northeast
African American women: 10/82 (12%) underwent GT Insurance type. Women with point-of-service plans were more likely to be tested than women with HMO plans
Recent diagnosis. Women diagnosed in 2007 were 3.8 times more likely to be tested than women diagnosed in 2004

Several studies conducted in non-U.S. settings have examined the uptake of genetic testing.[7-11] In studies examining the uptake of testing among at-risk relatives of carriers of BRCA1/BRCA2 pathogenic variants, uptake rates have averaged below 50% (range, 36%–48%), with higher uptake reported among female relatives than in male relatives. Other factors associated with higher uptake of testing were not consistently reported among studies but have most commonly included being a parent and wanting to learn information about a child’s risk.

Factors influencing uptake of genetic counseling and genetic testing

In reviews that have examined the cumulative evidence concerning the predictors of uptake of BRCA1/BRCA2 genetic testing, important predictors of testing uptake include older age, Ashkenazi Jewish (AJ) heritage, unmarried status, a personal history of breast cancer, and a family history of breast cancer. Studies recruiting participants in hospital settings had significantly higher recruitment rates than did studies recruiting participants in community settings. Studies that required an immediate decision to test, rather than allowing delayed decision making, tended to report higher uptake rates.[1] However, there is evidence that women diagnosed with breast cancer are equally satisfied with genetic counseling (including information received and strength and timing of physician recommendations for counseling), whether they received genetic counseling before or after their definitive surgery for breast cancer.[12] Another review [13] found that uptake of genetic testing for BRCA1/BRCA2 pathogenic variants was related to psychological factors (e.g., anxiety about breast cancer and perceived risk of breast cancer) and demographic and medical factors (e.g., history of breast cancer or ovarian cancer, presence of children, and higher number of affected first-degree relatives [FDRs]). Family members with a known BRCA1/BRCA2 pathogenic variant were more likely to pursue testing; those with more extensive knowledge of BRCA1/BRCA2 testing, heightened risk perceptions, beliefs that mammography would promote health benefit, and high intentions to undergo testing were more likely to follow through with testing.[14]

In a review of racial and ethnic differences that affect the uptake of BRCA1/BRCA2 testing, intention to undergo genetic testing in African American women was related to having at least one FDR with breast cancer or ovarian cancer, higher perceived risk of being a carrier, and less anticipatory guilt about the possibility of being a gene carrier.[15] A systematic review found that certain racial and ethnic minority groups, including African American and Hispanic individuals, had more negative views and greater concerns about genetic counseling and testing when compared with White individuals. African American and Hispanic individuals were more likely to believe genetic testing could be used to show their ethnic group was inferior to other groups. Additionally, African American and Hispanic individuals were found to have low awareness and knowledge about the importance of genetics in cancer, BRCA status, and genetic testing.[16]

Reasons cited for following through with testing included a desire to learn about a child's risk, to feel relief from uncertainty, to inform screening or risk-reducing surgery decisions, and to inform important life decisions such as marriage and childbearing.[14,17] Among African American women, the most important reason for testing included motivation to help other relatives decide on genetic testing.[15]

Physician recommendation may be another motivator for testing. In a retrospective study of 335 women considering genetic testing, 77% reported that they wanted the opinion of a genetics physician about whether they should be tested, and 49% wanted the opinion of their primary care provider.[18] However, there is some evidence of referral bias favoring those with a maternal family history of breast cancer or ovarian cancer. In a Canadian retrospective review of 315 patients, those with a maternal family history of breast cancer or ovarian cancer were 4.9 times (95% confidence interval, 3.6–6.7) more likely to be referred for a cancer genetics consultation by their physician than were those with a paternal family history (P < .001).[19] Studies have found that physicians may not adequately assess paternal family history [20] or may underestimate the significance of a paternal family history for genetic risk.[20-22] Other studies have shown that physician referral of patients who meet U.S. Preventive Services Task Force guidelines for BRCA genetic counseling has been suboptimal.[23]

The uptake of BRCA testing to inform surgical treatment decisions when offered appears to be high in research cohorts;[2,24] however, findings from other studies suggest that testing is underutilized in clinical practice to inform breast cancer treatment decisions.[6,25,26] Barriers to the use of BRCA testing to inform surgical treatment decisions, including lack of physician referral of newly diagnosed patients for genetic counseling, type of insurance coverage (such as Medicare or Medicaid), and challenges in the timing and coordination of testing, have been reported.[6,27-30] In a randomized trial that provided proactive rapid genetic counseling (delivery of genetic counseling prior to surgery) compared with usual care for patients with newly diagnosed breast cancer, results suggested that although genetic counseling uptake was higher in the intervention arm, this did not translate into higher rates of genetic testing, receipt of results before surgery, or bilateral mastectomy decisions.[31]

Insurance coverage

Insurance coverage is an important consideration for individuals deciding whether to undergo genetic testing. (Refer to the Insurance coverage section in the PDQ summary on Cancer Genetics Risk Assessment and Counseling for more information.)

Uptake of genetic counseling and genetic testing in diverse populations

Degree of uptake of genetic counseling and genetic testing in diverse populations

There are limited data on uptake of genetic counseling and testing among non-White populations, and further research will be needed to define factors influencing uptake in these populations.[32] The uptake of BRCA testing appears to vary across some racial and ethnic groups. A few studies have compared uptake rates between African American and White women.[4,33] In a case-control study of women who had been seen in a university-based primary care system, African American women with family histories of breast cancer or ovarian cancer were less likely to undergo BRCA1/BRCA2 testing than were White women who had similar histories.[33] In another study among breast cancer patients who were counseled about BRCA1/BRCA2 risk in a clinical setting, lower uptake was reported among African American women than among White women.[4]

Notably, the racial differences observed in these studies do not appear to be explained by factors related to cost, access to care, risk factors for carrying a BRCA1 or BRCA2 pathogenic variant, or differences in psychosocial factors, including risk perceptions, worry, or attitudes toward testing.

Factors influencing uptake of genetic counseling and genetic testing in diverse populations

Several studies have examined uptake or “acceptance” of BRCA testing among African American individuals enrolled in genetic research programs. Among study enrollees from an African American kindred in Utah, 83% underwent BRCA1 testing.[34] Age, perceived risk of being a carrier, and more extensive cancer knowledge predicted testing acceptance. Another study that recruited African American women through physician and community referrals reported a BRCA1/BRCA2 testing acceptance rate of 22%.[35] Predictors of test acceptance included having a higher probability of having a pathogenic variant, being married, and being less certain about one’s cancer risk. Finally, a third study that recruited at-risk African American women from an urban cancer screening clinic found that acceptors of BRCA testing were more knowledgeable about breast cancer genetics and perceived fewer barriers to testing, including negative emotional reactions, stigmatization concerns, and family-related guilt.[36] While these are independent predictors of genetic testing uptake, they do not explain the disparities in testing uptake across different ethnic groups. What may explain these differences are several attitudes and beliefs held about testing by individuals from diverse populations.

Work examining attitudes toward breast cancer genetic testing in Latino and African American populations indicates limited knowledge and awareness about testing but a generally receptive view once they are informed; in comparison with White populations, Latino and African American populations have relatively more concerns about testing.

For example, in a qualitative study with 51 Latino individuals unselected for risk status, important findings included the fact that participants were highly interested in genetic testing for inherited cancer susceptibility, despite very limited knowledge about genetics. One important barrier involved secrecy or embarrassment about family discussions of cancer and genetics, which could be addressed in intervention strategies.[37] Another qualitative study with 54 Latina women at risk of hereditary breast cancer showed that knowledge about BRCA1/BRCA2 counseling was low, although the women were interested in learning more about counseling to gain risk information for family members. Barriers to counseling included life demands, cost, and language issues.[38]

A telephone survey of 314 patients from an inner-city network of Pittsburgh, Pennsylvania, health centers, 50% of whom were African American, found that most participants (57%) (both African American and White participants) felt that genetic testing to evaluate disease risk was a good idea; however, more African American participants than White participants thought that genetic testing would lead to racial discrimination (37% vs. 22%, respectively) and that genetics research was unethical and tampered with nature (20% vs. 11%, respectively).[39] Finally, in a study of 222 women in Savannah, Georgia, where most had neither a personal history (70%) nor a family history (60%) of breast cancer, African American women (who comprised 26% of the sample) were less likely to be aware of breast cancer genes and genetic testing. Awareness was also related to higher income, higher education level, and having a family breast cancer history. However, 74% of the entire sample expressed willingness to be tested for breast cancer susceptibility.[40]

In a sample of 146 African American women meeting criteria for BRCA1/BRCA2 pathogenic variant testing, women born outside the United States reported higher levels of anticipated negative emotional reactions (e.g., fear, hopelessness, and lack of confidence that they could emotionally handle testing). Higher levels of breast cancer–specific distress were associated with anticipated negative emotional reactions, confidentiality concerns, and anticipated guilt regarding the family impact of breast cancer genetic testing.[41] A future orientation (e.g., "I often think about how my actions today will affect my health when I am older") was associated with overall perceived benefits of breast cancer genetic testing in this population (n = 140); however, future orientation was also found to be positively associated with family-related cons of testing, including family guilt and worry regarding the impact of testing on the family.[42]

There are racial differences in provider discussion and patient uptake of genetic testing for variants in BRCA1/BRCA2. A study of women aged 18 to 64 years and diagnosed with invasive breast cancer between 2007 and 2009 found that, even after adjusting for pathogenic variant risk, African American women were less likely to report having received a physician recommendation for genetic testing. There was no difference across all races in concerns that BRCA1/BRCA2 testing was too expensive and only minimal differences in testing attitudes or insurance concerns were found, none of which influenced testing uptake.[43] A study of breast or ovarian cancer survivors (N = 50) eligible for BRCA1/BRCA2 genetic testing found that 48% were referred for genetic counseling and testing and/or had undergone genetic testing. Individuals with higher breast cancer genetics knowledge and higher self-efficacy were more likely to have engaged in genetic counseling and testing.[44] In a study of women with invasive breast cancer diagnosed before age 50 years between 2009 and 2012 who were identified through the Florida Cancer Data System state registry and eligible for BRCA1/BRCA2 genetic testing on the basis of existing guidelines, African American individuals were less likely to report a discussion with their health care provider and undergo genetic testing.[45] The same study found similar overall testing rates in Hispanic (61%) and non-Hispanic (65%) White individuals. However, testing rates were lower among Hispanic individuals who spoke primarily Spanish at home (50% Spanish speaking vs. 69% English speaking; P = .0009), and in general, Hispanic individuals were less likely to have been referred for genetic testing.[46] However, this finding is not consistent across all studies. In a study of women aged 20 to 79 years with ductal carcinoma in situ or invasive breast cancer identified through the Surveillance, Epidemiology, and End Results (SEER) registry in Georgia and Los Angeles County, all eligible for BRCA1/BRCA2 genetic testing on the basis of existing guidelines, no ethnic differences were detected in receipt of genetic counseling or physician-directed discussion about genetic testing.[30]

Factors associated with declining genetic counseling and testing

There is evidence that primary reasons for declining testing involves being childless, which reduces any family motivations for testing; and concerns about the negative ramifications of testing, including difficulty retaining insurance or concerns about personal health.

Limited data are available about the characteristics of at-risk individuals who decline to be tested or have never been tested. It is difficult to access samples of test decliners because they may be reluctant to participate in research studies. Studies of genetic testing uptake are difficult to compare because people may decline at different points and with different amounts of pretest education and counseling. One study found that 43% of affected and unaffected individuals from hereditary breast/ovarian cancer families who completed a baseline interview regarding testing declined to be tested. Most individuals who declined testing chose not to participate in educational sessions. Decliners were more likely to be male and be unmarried and have fewer relatives with breast cancer. Decliners who had high levels of cancer-related stress had higher levels of depression. Decliners lost to follow-up were significantly more likely to be affected with cancer.[47]

Another study looked at a small number (n = 13) of women decliners who carried a 25% to 50% probability of harboring a BRCA pathogenic variant; these nontested women were more likely to be childless and to have higher levels of education. This study showed that most women decided not to undergo the test after serious deliberation about the risks and benefits. Satisfaction with frequent surveillance was given as one reason for nontesting by most of these women.[48] Other reasons for declining included having no children and becoming acquainted with breast/ovarian cancer in the family relatively early in their lives.[47,48]

A third study evaluated characteristics of 34 individuals who declined BRCA1/BRCA2 testing in a large multicenter study in the United Kingdom. Decliners were younger than a national sample of test acceptors, and female decliners had lower mean scores on a measure of cancer worry. Although 78% of test decliners/deferrers felt that their health was at risk, they reported that learning about their BRCA1/BRCA2 pathogenic variant status would cause them to worry about the following:

  • Their children's health (76%).
  • Their life insurance (60%).
  • Their own health (56%).
  • Loss of their job (5%).
  • Receiving less screening if they did not carry a BRCA1/BRCA2 pathogenic variant (62%).

Apprehension about the impact of the test result was a more important factor in the decision to decline testing than were concrete burdens such as time required to travel to a genetics clinic and time spent away from work, family, and social obligations.[49] In 15% (n = 31) of individuals from 13 hereditary breast and ovarian cancer families who underwent genetic education and counseling and declined testing for a documented pathogenic variant in the family, positive changes in family relationships were reported—specifically, greater expressiveness and cohesion—compared with those who pursued testing.[50]

Genetic counseling and testing in children

Testing for BRCA1/BRCA2 pathogenic variants has been almost universally limited to adults older than 18 years. The risks of testing children for adult-onset disorders, such as breast and ovarian cancers, as inferred from developmental data on children’s medical understanding and ability to provide informed consent, have been outlined in several reports.[51-54]

Studies suggest that individuals who have undergone BRCA1/BRCA2 genetic testing or who are adult offspring of individuals who have had testing are generally not in favor of testing minors.[55,56] Although the data are limited, research suggests that males, pathogenic variant noncarriers, and those whose mothers did not have personal histories of breast cancer may be more likely to favor genetic testing in minors in general.[55] Of those who had minor children at the time the study was conducted, only 17% stated a preference for having their own children tested. Concerns regarding testing of minors included psychological risks and insufficient maturity. Potential benefits included the ability to influence health behaviors.[56]

No data exist on the testing of children for BRCA1/BRCA2 pathogenic variants, although some researchers believe it is necessary to test the validity of assumptions underlying the general prohibition of testing children for genetic variants associated with breast and ovarian cancers and other adult-onset diseases.[57-59] In one study, 20 children (aged 11–17 y) of a selected group of mothers undergoing genetic testing (80% of whom previously had breast cancer and all of whom had discussed BRCA1/BRCA2 testing with their children) completed self-report questionnaires on their health beliefs and attitudes toward cancer, feelings related to cancer, and behavioral problems.[60] Ninety percent of children thought they would want cancer risk information as adults; half worried about themselves or a family member developing cancer. There was no evidence of emotional distress or behavioral problems.

What People Bring to Genetic Testing: Impact of Risk Perception, Health Beliefs, and Personality Characteristics

The emerging literature in this area suggests that risk perceptions, health beliefs, psychological status, and personality characteristics are important factors in decision making about breast/ovarian cancer genetic testing. Many women presenting at academic centers for BRCA1/BRCA2 testing arrive with a strong belief that they have a pathogenic variant, having decided they want genetic testing, but possessing little information about the risks or limitations of testing.[61] Most mean scores of psychological functioning at baseline for subjects in genetic counseling studies were within normal limits.[62] Nonetheless, a subset of subjects in many genetic counseling studies present with elevated anxiety, depression, or cancer worry.[63,64] Identification of these individuals is essential to prevent adverse outcomes. In a study of 205 women pursuing genetic counseling, interactions among cancer worry, breast cancer risk perception, and perceived severity of having a breast cancer genetic variant were found such that those with high worry, high breast cancer risk perception, and low perceived severity were twice as likely to follow through with BRCA1/BRCA2 testing than others.[65]

A general tendency to overestimate inherited risk of breast and ovarian cancer has been noted in at-risk populations,[66-69] in cancer patients,[67,70,71] in spouses of breast and ovarian cancer patients,[72] and among women in the general population.[73-75] but underestimation of breast cancer risk in higher-risk and average-risk women also has been reported.[76] This overestimation may encourage a belief that BRCA1/BRCA2 genetic testing will be more informative than it is currently thought to be. Some evidence exists that even counseling does not dissuade women at low to moderate risk from the belief that BRCA1 testing could be valuable.[32] Overestimation of both breast and ovarian cancer risk has been associated with nonadherence to physician-recommended screening practices.[77,78] A meta-analysis of 12 studies of outcomes of genetic counseling for breast/ovarian cancer showed that counseling improved the accuracy of risk perception.[79]

Women appear to be the prime communicators within families about the family history of breast cancer.[80] Higher numbers of maternal versus paternal transmission cases are reported,[81] likely due to family communication patterns, to the misconception that breast cancer risk can only be transmitted through the mother, and to the greater difficulty in recognizing paternal family histories because of the need to identify more distant relatives with cancer. In an analysis of 2,505 women participating in the Family Healthware Impact Trial,[82] not only was evidence of underreporting of paternal family history identified, but also women reported a lower level of perceived breast cancer risk with a paternal versus maternal breast cancer family history.[83] Physicians and counselors taking a family history are encouraged to elicit paternal and maternal family histories of breast, ovarian, or other associated cancers.[80]

The accuracy of reported family history of breast or ovarian cancer varies; some studies found levels of accuracy above 90%,[84,85] with others finding more errors in the reporting of cancer in second-degree or more distant relatives [86] or in age of onset of cancer.[87] Less accuracy has been found in the reporting of cancers other than breast cancer. Ovarian cancer history was reported with 60% accuracy in one study compared with 83% accuracy in breast cancer history.[88] Providers should be aware that there are a few published cases of Munchausen syndrome in reporting of false family breast cancer history.[89] Much more common is erroneous reporting of family cancer history due to unintentional errors or gaps in knowledge, related in some cases to the early death of potential maternal informants about cancer family history.[80] (Refer to the Taking a Family History section of the Cancer Genetics Risk Assessment and Counseling summary for more information.)

Targeted written,[90,91] video, CD-ROM, interactive computer programs and websites,[92-99] and culturally targeted educational materials [100-102] may be effective and efficient methods of increasing knowledge about the pros and cons of genetic testing. Such supplemental materials may allow more efficient use of the time allotted for pretest education and counseling by genetics and primary care providers and may discourage individuals without appropriate indication of risk from seeking genetic testing.[90]

Genetic Counseling for Hereditary Predisposition to Breast Cancer

Counseling for breast cancer risk typically involves individuals with family histories that are potentially attributable to BRCA1 or BRCA2. It also, however, may include individuals with family histories of Li-Fraumeni syndrome, ataxia-telangiectasia, Cowden syndrome, or Peutz-Jeghers syndrome.[103] (Refer to the High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes section of this summary for more information.)

Management strategies for carriers may involve decisions about the nature, frequency, and timing of screening and surveillance procedures, chemoprevention, risk-reducing surgery, and use of hormone replacement therapy (HRT). The utilization of breast conservation and radiation as cancer therapy for women who are carriers may be influenced by knowledge of pathogenic variant status. (Refer to the Clinical Management of Carriers of BRCA Pathogenic Variants section of this summary for more information.)

Counseling also includes consideration of related psychosocial concerns and discussion of planned family communication and the responsibility to warn other family members about the possibility of having an increased risk of breast, ovarian, and other cancers. Data suggest that individual responses to being tested as adults are influenced by the results status of other family members.[104,105] Management of anxiety and distress are important not only as quality-of-life factors, but also because high anxiety may interfere with the understanding and integration of complex genetic and medical information and adherence to screening.[106-108] Formal, objective evaluation of these outcomes are well documented. (Refer to the Emotional Outcomes and Behavioral Outcomes sections of this summary for more information.)

Published descriptions of counseling programs for BRCA1 (and subsequently for BRCA2) testing include strategies for gathering a family history, assessing eligibility for testing, communicating the considerable volume of relevant information about breast/ovarian cancer genetics and associated medical and psychosocial risks and benefits, and discussion of specialized ethical considerations about confidentiality and family communication.[109-116] Participant distress, intrusive thoughts about cancer, coping style, and social support were assessed in many prospective testing candidates. The psychosocial outcomes evaluated in these programs have included changes in knowledge about the genetics of breast/ovarian cancer after counseling, risk comprehension, psychological adjustment, family and social functioning, and reproductive and health behaviors.[117] A Dutch study of communication processes and satisfaction levels of consultands going through cancer genetic counseling for inherited cancer syndromes indicated that asking more medical questions (by the counselor), providing more psychosocial information, and longer eye contact by the counselor were associated with lower satisfaction levels. The provision of medical information by the counselor was most highly related to satisfaction and perception that needs have been fulfilled.[118]

Many of the psychosocial outcome studies involve specialized, highly selected research populations, some of which were utilized to map and clone BRCA1 and BRCA2. One such example is K2082, an extensively studied kindred of more than 800 members of a Utah Mormon family in which a BRCA1 pathogenic variant accounts for the observed increased rates of breast and ovarian cancer. A study of the understanding that members of this kindred have about breast/ovarian cancer genetics found that, even in breast cancer research populations, there was incomplete knowledge about associated risks of colon and prostate cancer, the existence of options for RRM and RRSO, and the complexity of existing psychosocial risks.[109] A meta-analysis of 21 studies found that genetic counseling was effective in increasing knowledge and improved the accuracy of perceived risk. Genetic counseling did not have a statistically significant long-term impact on affective outcomes including anxiety, distress, or cancer-specific worry and the behavioral outcome of cancer surveillance activities.[62] These prospective studies, however, were characterized by a heterogeneity of measures of cancer-specific worry and inconsistent findings in effects of change from baseline.[62]

Emotional Outcomes

Although there were initial concerns about the possibility of adverse emotional consequences from BRCA testing, most studies conducted over the years have shown low levels of psychological distress among both carriers and noncarriers, particularly over the longer term.[119-121] In a meta-analysis examining cancer-specific distress over short (0–4 weeks), moderate (5–24 weeks), and long (25–52 weeks) periods of time since the receipt of testing results, carriers were found to demonstrate increased levels of distress shortly after receiving results, with levels returning to baseline within moderate and long periods of time.[119] In contrast, noncarriers and those with inconclusive results showed reduced levels of distress over time.[119,122] Psychological distress patterns were found to vary as a function of several factors, including the cancer history of the individual and the country within which the study was conducted. Carriers with a personal history of cancer experienced small decreases in distress over time, whereas no changes were observed among carriers without a personal history of cancer. Among individuals with inconclusive results, greater decreases in distress were observed among those without a cancer history than among those with a cancer history. Among noncarriers, those in the United States experienced significantly greater decreases in psychological distress than noncarriers from Europe and Australia. A study conducted in Austria noted that certain subgroups of consultands experienced greater distress, including those who were older, had a more recent cancer diagnosis, or those who had received counseling but declined BRCA testing.[123]

Several studies have reported on emotional outcomes over longer follow-up periods (i.e., greater than 12 months after disclosure) than those reported in the meta-analysis described above.[119] In a U.K. study, cancer-related worry did not differ between carriers and noncarriers at 3 years of follow-up.[124] Two U.S.-based studies published since the meta-analytic review [119] have reported similar findings among women who were surveyed more than 3 years after receipt of BRCA test results.[125,126] In a cross-sectional study,[125] 167 women who were surveyed more than 4 years after receiving BRCA test results reported low levels of genetic testing–specific concerns, as measured using the Multidimensional Impact of Cancer Risk Assessment Scale.[127] In multivariate regression models, carriers of pathogenic variants were significantly more likely to experience distress than were noncarriers. In a second study,[126] 464 women were followed prospectively for a median of 5 years (range, 3.4–9.1 y) after testing. Among both affected and unaffected participants, BRCA carriers reported significantly higher levels of distress, uncertainty (affected only), perceived stress (affected only), and lower positive testing experiences (unaffected only) than women who received negative results for a known pathogenic variant in the family.[126] Although both studies [125,126] reported greater distress among BRCA carriers than among noncarriers, the level of distress was not reflective of clinically significant dysfunction.

Although most studies have reported that a positive BRCA test result has a relatively minimal impact on psychological distress, many of these studies were conducted among families with a strong family history of breast or ovarian cancer who underwent extensive pretest genetic counseling. Therefore, emotional responses may not generalize to individuals who test under different contexts. For example, individuals who are tested with population BRCA screening may not have a family history of cancer.[128-130] Although pretest genetic counseling is recommended, this is not always done when genetic testing is ordered by nongenetic providers [131] or directly through commercial companies.[132,133]

For example, in a Canadian study of 2,080 Jewish women who participated in a population-based genetic screening study to test for three BRCA pathogenic variants common in families of Jewish heritage, women were not offered in-person genetic counseling but were given a pamphlet on genetic testing for BRCA1/BRCA2 before they provided a DNA sample. One year after genetic testing, women who were positive for a pathogenic variant (n = 18) showed significant increases in cancer-specific distress, whereas no changes in distress were observed among women who were negative for a pathogenic variant.[129] The mean distress score on the Impact of Event Scale for the 18 women with a known pathogenic variant was 25.3 (range, 2–51); 10 of 18 women (56%) scored within moderate (26–43) (n = 7) or severe (44+) (n = 3) ranges. It is unclear from this study whether the increase in distress observed at 1 year of follow-up was due to the lack of in-person genetic counseling, or whether the lower levels of distress at baseline observed were because the women in the study were low risk but eligible for testing because of their ancestry. A follow-up study with this cohort found that distress decreased between 1 to 2 years after testing and that changes in distress varied by risk-reduction options undertaken by carriers. Specifically, those who had undergone risk-reducing mastectomy or oophorectomy experienced significant decreases in distress compared with those who did not have either surgery.[130] Another smaller qualitative study also supports these findings.[134]

Similarly, the impact of direct-to-consumer (DTC) BRCA testing through commercial companies requires further evaluation. Case studies have reported adverse emotional responses after receipt of a positive BRCA result from DTC genetic testing, suggesting the need for further evaluation of the emotional outcomes of women undergoing genetic testing through commercial companies.[132,133] Only one study, conducted by a commercial company, has attempted to evaluate the impact of BRCA testing in this context.[135] A total of 32 individuals (16 women and 16 men) who tested positive for one of three BRCA founder pathogenic variants common in Ashkenazi Jews completed semi-structured interviews. None of the carriers reported extreme anxiety, although some experienced moderate anxiety (13%) or initial disappointment and anxiety that dissipated over time (28%). These findings should be interpreted with caution given that only 24% (32 of 136) of invited carriers of BRCA pathogenic variants participated in the study, raising concerns about selection bias.

Despite evidence of a short-term increase in distress after the receipt of genetic testing results, any adverse responses to a positive carrier status dissipate within 12 months.[119] Additional research is needed to examine emotional outcomes for those who are not provided genetic counseling before testing.[131]

Emotional outcomes in newly diagnosed breast cancer patients

It is increasingly common for women with breast cancer to pursue genetic counseling and testing at the time of diagnosis to assist with treatment decision making. (Refer to the Benefits of offering genetic testing at the time of cancer diagnosis section in the Introduction section of this summary for more information.) Although concerns have been raised about the adverse psychological implications of offering rapid genetic counseling and testing between diagnosis and surgery,[136,137] other studies,[138-140] including a randomized trial,[141] have provided evidence indicating no additional adverse psychological effects in newly diagnosed breast cancer patients. One randomized controlled trial found that patients undergoing rapid genetic counseling and testing felt more actively involved in treatment decision making than those receiving standard care.[142] However, qualitative research on 20 newly diagnosed breast cancer patients found that some subgroups of these patients may have more difficulty coping with BRCA test results, such as carriers who have no family history of cancer; those who do not have an affected relative with whom they can identify; and higher risk women who receive uninformative negative BRCA results.[136]

Family Effects

Family communication about genetic testing and hereditary risk

Family communication about genetic testing for cancer susceptibility, and specifically about the results of BRCA1/BRCA2 genetic testing, is complex. Gender appears to be an important variable in family communication and psychological outcomes. Studies have documented that female carriers are more likely to disclose their status to other family members (especially sisters and children aged 14–18 y) [143] than are male carriers.[143,144] Among males, noncarriers were more likely than carriers to tell their sisters and children the results of their tests. BRCA1/BRCA2 carriers who disclosed their results to sisters had a slight decrease in psychological distress, compared with a slight increase in distress for carriers who chose not to tell their sisters. One study found that men reported greater difficulty disclosing a known pathogenic variant to family members than women (90% vs. 70%).[145]

Family communication of BRCA1/BRCA2 test results to relatives is another factor affecting participation in testing. There have been more studies of communication with FDRs and second-degree relatives than with more distant family members. Studies have investigated the process and content of communication among sisters about BRCA1/BRCA2 test results.[146,147] Study results suggest that both carriers of pathogenic variants [146] and women with uninformative results [146,147] communicate with sisters to provide them with genetic risk information. Similar findings were reported in women with uninformative results disclosing test results to their daughters.[147] Among relatives with whom genetic test results were not discussed, the most important reason given was that the affected women were not close to their relatives [146] or had a poor relationship with them.[147] Studies found that women with a BRCA pathogenic variant more often shared their results with their mother and adult sisters and daughters than with their father and adult brothers and sons.[80,148-151] A study that evaluated communication of test results to FDRs at 4 months postdisclosure found that women aged 40 years or older were more likely to inform their parents of test results compared with younger women. Participants also were more likely to inform brothers of their results if the BRCA pathogenic variant was inherited through the paternal line.[149] Another study found that disclosure was limited mainly to FDRs, and dissemination of information to distant relatives was problematic.[152] Age was a significant factor in informing distant relatives with younger patients being more willing to communicate their genetic test result.[146,148,152] Additionally, one study found that lower genetic worry, higher interest in genomic information, carrying a BRCA1 or BRCA2 pathogenic variant, or having never been married was associated with communication to more family members.[144] In contrast, a longer time interval since diagnosis was associated with communication to fewer family members.[144]

A few in-depth qualitative studies have looked at issues associated with family communication about genetic testing. Although the findings from these studies may not be generalizable to the larger population of at-risk individuals, they illustrate the complexity of issues involved in conveying hereditary cancer risk information in families.[153] On the basis of 15 interviews conducted with women attending a familial cancer genetics clinic, the authors concluded that while women felt a sense of duty to discuss genetic testing with their relatives, they also experienced conflicting feelings of uncertainty, respect, and isolation. Decisions about whom in the family to inform and how to inform them about hereditary cancer and genetic testing may be influenced by tensions between women's need to fulfill social roles and their responsibilities toward themselves and others.[153] Another qualitative study of 21 women who attended a familial breast and ovarian cancer genetics clinic suggested that some women may find it difficult to communicate about inherited cancer risk with their partners and with certain relatives, especially brothers, because of their own fears and worries about cancer.[151] This study also suggested that how genetic risk information is shared within families may depend on the existing norms for communicating about cancer in general. For example, family members may be generally open to sharing information about cancer with each other, may selectively avoid discussing cancer information with certain family members to protect themselves or other relatives from negative emotional reactions, or may ask a specific relative to act as an intermediary to disclosure of information to other family members.[154] The potential importance of individuals outside the family, such as friends, as both confidantes about inherited cancer risk information and as sources of support for coping with this information was also noted in the study.[151]

A study of 31 mothers with a documented BRCA pathogenic variant explored patterns of dissemination to children.[155] Of those who chose to disclose test results to their children, age of offspring was the most important factor. Fifty percent of the children who were told were aged 20 to 29 years and slightly more than 25% of the children were aged 19 years or younger. Sons and daughters were notified in equal numbers. More than 70% of mothers informed their children within a week of learning their test result. Ninety-three percent of mothers who chose not to share their results with their children indicated that it was because their children were too young. These findings were consistent with three other studies showing that children younger than 13 years were less likely to be informed about test results compared with older children.[149,156,157] Another study of 187 mothers undergoing BRCA1/BRCA2 testing evaluated their need for resources to prepare for a facilitated conversation about sharing their BRCA1/BRCA2 testing results with their children. Seventy-eight percent of mothers were interested in three or more resources, including literature (93%), family counseling (86%), talk to prior participants (79%), and support groups (54%).[156]

A longitudinal study of 153 women self-referred for genetic testing for BRCA1 and BRCA2 pathogenic variants and 118 of their partners evaluated communication about genetic testing and distress before testing and at 6 months posttesting.[158] The study found that most couples discussed the decision to undergo testing (98%), most test participants felt their partners were supportive, and most women disclosed test results to their partners (97%, n = 148). Test participants who felt their partners were supportive during pretest discussions experienced less distress after disclosure, and partners who felt more comfortable sharing concerns with test participants pretest experienced less distress after disclosure. Six-month follow-up revealed that 22% of participants felt the need to talk about the testing experience with their partners in the week before the interview. Most participants (72%, n = 107) reported comfort in sharing concerns with their partners, and 5% (n = 7) reported relationship strain as a result of genetic testing. In couples in which the woman had a positive genetic test result, more relationship strain, more protective buffering of their partners, and more discussion of related concerns were reported than in couples in which the woman had a true-negative or uninformative result.[158]

A study of 561 FDRs of women who had undergone BRCA1/BRCA2 genetic testing found that 22% of FDRs did not recall being informed of the genetic test results despite the women reporting that the results had been shared.[159] Men were less likely to recall receiving the results (P > .001). Of those with recall about receiving the test results, 10.5% of FDRs did not recall the findings. For those with recall of the results, 17.9% of FDRs had an interpretation that was discordant with the correct results. Accuracy of test results recall was greater for informative test results (those that were either true positive or true negative) (P = .029). However, regardless of the test results, FDRs perceived the cancer risk to be higher before they learned of the findings than after (74% and 53% of FDRs reported that they believed their risk for cancer was greater than average before and after hearing test results, respectively).

There is a small but growing body of literature regarding psychological effects in men who have a family history of breast cancer and who are considering or have had BRCA testing. A qualitative study of 22 men from 16 high-risk families in Ireland revealed that more men in the study with daughters were tested than men without daughters. These men reported little communication with relatives about the illness, with some men reporting being excluded from discussion about cancer among female family members. Some men in the study also reported actively avoiding open discussion with daughters and other relatives.[160] In contrast, a study of 59 men testing positive for a BRCA1/BRCA2 pathogenic variant found that most men participated in family discussions about breast and/or ovarian cancer. However, fewer than half of the men participated in family discussions about risk-reducing surgery. The main reason given for having BRCA testing was concern for their children and a need for certainty about whether they could have transmitted the pathogenic variant to their children. In this study, 79% of participating men had at least one daughter. Most of these men described how their relationships had been strengthened after receipt of BRCA results, helping communication in the family and greater understanding.[161] Men in both studies expressed fears of developing cancer themselves. Irish men especially reported fear of cancer in sexual organs.

Family functioning

One study assessed 212 individuals from 13 hereditary breast and ovarian cancer families who received genetic counseling and were offered BRCA1/BRCA2 testing for documented pathogenic variants in the family. Individuals who were not tested were found 6 to 9 months later to have significantly greater increases in family expressiveness and cohesiveness compared with those who were tested. Individuals who were randomly assigned to a client-centered versus problem-solving genetic counseling intervention had a significantly greater reduction in conflict, regardless of the test decision.[50]

Partners of high-risk women

Many studies have looked at the psychological effects in women of having a high risk of developing cancer, either on the basis of carrying a BRCA1/BRCA2 pathogenic variant or having a strong family history of cancer. Some studies have also examined the effects on the partners of such women.

A Canadian study assessed 59 spouses of women found to have a BRCA1/BRCA2 pathogenic variant. All were supportive of their spouses’ decision to undergo genetic testing and 17% wished they had been more involved in the genetic testing process. Spouses who reported that genetic testing had no impact on their relationship had long-term relationships (mean duration 27 years). Forty-six percent of spouses reported that their major concern was of their partner dying of cancer. Nineteen percent were concerned their spouse would develop cancer and 14% were concerned their children would also be carriers of BRCA1/BRCA2 pathogenic variants.[162]

In a U.S. study, 118 partners of women who underwent genetic testing for pathogenic variants in BRCA1 and BRCA2 completed a survey before testing and then again 6 months after result disclosure. At 6 months, only 10 partners reported that they had not been told of the test result. Ninety-one percent reported that the testing had not caused strain on their relationship. Partners who were comfortable sharing concerns before testing experienced less distress after testing. Protective buffering was not found to impact distress levels of partners.[158]

An Australian study of 95 unaffected women at high risk of developing breast and/or ovarian cancer (13 carriers of pathogenic variants and 82 with unknown variant status) and their partners showed that although the majority of male partners had distress levels comparable to a normative population sample, 10% had significant levels of distress that indicated the need for further clinical intervention. Men with a high monitoring coping style and greater perceived breast cancer risk for their wives reported higher levels of distress. Open communication between the men and their partners and the occurrence of a cancer-related event in the wife’s family in the last year were associated with lower distress levels. When men were asked what kind of information and support they would like for themselves and their partners, 57.9% reported that they would like more information about breast and ovarian cancer, and 32.6% said they would like more support in dealing with their partner's risk. Twenty-five percent of men had suggestions on how to improve services for partners of high-risk women, including strategies on how to best support their partner, greater encouragement from health care professionals to attend appointments, and meeting with other partners.[163]

A review of this literature reported that the BRCA testing process may be distressing for male partners, particularly for those with spouses identified as carriers. Male partner distress appears to be associated with their beliefs about the woman’s breast cancer risk, lack of couple communication, and feelings of alienation from the testing process.[164]

At-risk males

A review of the literature on the experiences of males in families with a known BRCA1 and BRCA2 pathogenic variant reported that while the data are limited, men from variant-positive families are less likely than females to participate in communication regarding genetics at every level, including the counseling and testing process. Men are less likely to be informed of genetic test results received by female relatives, and most men from these families do not pursue their own genetic testing.[165]

A study of Dutch men at increased risk of having inherited a BRCA1 pathogenic variant reported a tendency for the men to deny or minimize the emotional effects of their risk status, and to focus on medical implications for their female relatives. Men in these families, however, also reported considerable distress in relation to their female relatives.[166] In another study of male psychological functioning during breast cancer testing, 28 men belonging to 18 different high-risk families (with a 25% or 50% risk of having inherited a BRCA1/BRCA2 pathogenic variant) participated. The study purpose was to analyze distress in males at risk of carrying a BRCA1/BRCA2 pathogenic variant who applied for genetic testing. Of the men studied, most had low pretest distress; scores were lowest for men who were optimistic or who did not have daughters. Most carriers of pathogenic variants had normal levels of anxiety and depression and reported no guilt, though some anticipated increased distress and feelings of responsibility if their daughters developed breast or ovarian cancer. None of the noncarriers reported feeling guilty.[167] In one study,[161] adherence to recommended screening guidelines after testing was analyzed. In this study, more than half of male carriers of pathogenic variants did not adhere to the screening guidelines recommended after disclosure of genetic test results. These findings are consistent with those for female carriers of BRCA1/BRCA2 pathogenic variants.[161,168]

A multicenter U.K. cohort study examined prospective outcomes of BRCA1/BRCA2 testing in 193 individuals, of which 20% were men aged 28 to 86 years. Men’s distress levels were low, did not differ among carriers and noncarriers, and did not change from baseline (before genetic testing) to the 3-year follow-up. Twenty-two percent of male carriers of pathogenic variants received colorectal cancer screening and 44% received prostate cancer screening;[124] however, it is unclear whether men in this study were following age-appropriate screening guidelines.

Children

Several studies have explored communication of BRCA test results to at-risk children. Across all studies, the rate of disclosure to children ranging in age from 4 to 25 years is approximately 50%.[148,149,152,156,169-172] In general, age of offspring was the most important factor in deciding whether to disclose test results. In one study of 31 mothers disclosing their BRCA test results, 50% of the children who were informed of the results were aged 20 to 29 years and slightly more than 25% of the children were aged 19 years or younger. Sons and daughters were notified in equal numbers.[155] Similarly, in another study of 42 female carriers of BRCA pathogenic variants, 83% of offspring older than age 18 years were told of the results, while only 21% of offspring aged 13 years or younger were told.[156]

Several studies have also looked at the timing of disclosure to children after parents receive their test results. Although the majority of children were told within a week to several months after results disclosure,[149,155,156] some parents chose to delay disclosure.[156] Reasons for delaying disclosure included waiting for the child to get older, allowing time for the parent to adjust to the information, and waiting until results could be shared in person (in the case of adult children living away from home).[156]

In one study, participants who told children younger than 13 years about their carrier status had increased distress, and those who did not tell their young children experienced a slight decrease in distress. Communication with young children was found to be influenced by developmental variables such as age and style of parent/child communication.[171]

One study looked at the reaction of children to results disclosure or the effect on the parent-child relationship of communicating the results.[156] With regard to offspring’s understanding of the information, almost half of parents from one study reported that their child did not appear to understand the significance of a positive test result, although older children were reported to have a better understanding. This same study also showed that 48% of parents reported at least one negative reaction in their child, ranging from anxiety or concern (22%) to crying and fear (26%). It should be noted, however, that in this study children's level of understanding and reactions to the test result were measured qualitatively and based only on the parents' perception. Also, given the retrospective design of the study, there was a potential for recall bias. There were no significant differences in emotional reaction depending on age or gender of the child. Lastly, 65% of parents reported no change in their relationship with their child, while 5 parents (22%) reported a strengthening of their relationship.

Interestingly, a large multicenter study of 869 mother-daughter pairs (the daughters were aged 6 to 13 y) found that girls with a family history of breast cancer or a familial BRCA1/BRCA2 pathogenic variant (BCFH+) compared with those without such family histories had better psychosocial adjustment by maternal report.[173] However, based on a combination of maternal report and direct assessment of girls aged 10 to 13 years, BCFH+ girls experienced greater breast cancer–specific distress and a higher perceived risk of breast cancer than their peers without such family histories. Moreover, higher daughter distress was associated with higher maternal distress. A similarly designed study in older girls, aged 11 to 19 years, found that higher breast cancer–specific distress in daughters was associated with perceived risk and maternal distress. This older age group had higher self-esteem than did their peers without a family history of breast cancer.[174]

Another study of 187 mothers undergoing BRCA1/BRCA2 testing evaluated their need for resources to prepare for a facilitated conversation about sharing their BRCA1/BRCA2 testing results with their children. Seventy-eight percent of mothers were interested in three or more resources, including literature (93%), family counseling (86%), talking to prior participants (79%), and support groups (54%).[175]

Testing for BRCA1/BRCA2 has been almost universally limited to adults older than 18 years. The risks of testing children for adult-onset disorders (such as breast and ovarian cancer), as inferred from developmental data on children’s medical understanding and ability to provide informed consent, have been outlined in several reports.[51-54] Surveys of parental interest in testing children for adult-onset hereditary cancers suggest that parents are more eager to test their children than to be tested themselves for a breast cancer gene, suggesting potential conflicts for providers.[176,177] In a general population survey in the United States, 71% of parents said that it was moderately, very, or extremely likely that if they carried a breast-cancer predisposing pathogenic variant, they would test a 13-year-old daughter now to determine her breast cancer gene status.[176] To date, no data exist on the testing of children for BRCA1/BRCA2, though some researchers believe it is necessary to test the validity of assumptions underlying the general prohibition of testing of children for breast/ovarian cancer and other adult-onset disease genes.[57-59] In one study, 20 children (aged 11–17 y) of a selected group of mothers undergoing genetic testing (80% of whom previously had breast cancer and all of whom had discussed BRCA1/BRCA2 testing with their children) completed self-report questionnaires on their health beliefs and attitudes toward cancer, feelings related to cancer, and behavioral problems.[60] Ninety percent of children thought they would want cancer risk information as adults; half worried about themselves or a family member developing cancer. There was no evidence of emotional distress or behavioral problems. Another study by this group [171] found that 1 month after disclosure of BRCA1/BRCA2 genetic test results, 53% of 42 enrolled mothers of children aged 8 to 17 years had discussed their result with one or more of their children. Age of the child rather than pathogenic variant status of the mother influenced whether they were told, as did family health communication style.

Prenatal diagnosis and preimplantation genetic testing

The possibility of transmitting a pathogenic variant to a child may pose a concern to families affected by hereditary breast and ovarian cancer (HBOC),[178] perhaps to the extent that some carriers may avoid childbearing.[179,180] These concerns also may prompt women to consider using prenatal testing methods to help reduce the risk of transmission.[178,181] Prenatal diagnosis is an encompassing term used to refer to any medical procedure conducted to assess the presence of a genetic disorder in a fetus. Methods include amniocentesis and chorionic villous sampling (CVS).[182,183] Both procedures carry some risk of miscarriage and some evidence suggests fetal defects may result from using these tests.[182,183] Moreover, discovering the fetus is a carrier for a genetic defect may impose a difficult decision for couples regarding pregnancy continuation or termination. An alternative to these tests is preimplantation genetic testing (PGT), a procedure used to test fertilized embryos for genetic disorders before uterine implantation,[178,184,185] thereby avoiding the potential dangers associated with amniocentesis and CVS and the decision to terminate a pregnancy. Using the information obtained from the genetic testing, potential parents can decide whether or not to implant. PGT can be used to detect pathogenic variants in hereditary cancer predisposing genes, including BRCA.[178,181]

In the United States, a series of studies has evaluated awareness, interest (e.g., would consider using PGT), and attitudes related to PGT among members of Facing Our Risk of Cancer Empowered (FORCE), an advocacy organization focused on individuals at increased risk of HBOC.[178,181,186] The first study was a Web-based survey of 283 members,[178] the second included 205 attendees of the 2007 annual FORCE conference,[181] and the third was a Web-based survey of 962 members.[186,187] These studies have documented low levels of awareness, with 20% to 32% of study respondents reporting having heard of PGT before study participation.[181,186] With respect to interest in PGT, the first study [178] found only 13% of women would be likely to use PGT, whereas, 33% of respondents in the subsequent FORCE studies reported that they would consider using PGT.[181,186] In the third FORCE-based study (n = 962),[186] multivariable analysis revealed PGT interest was associated with the desire to have more children, having previously had any prenatal genetic test, and previous awareness of PGT. Attitudinal predictors of interest in PGT included agreement that others at risk of HBOC should be offered PGT; the belief that PGT is acceptable for individuals at risk of HBOC; the belief that PGT information should be given to individuals at risk of HBOC; and endorsement of PGT benefits of having children without genetic variants and eliminating genetic diseases. Conversely, those who indicated that PGT was “too much like playing God” and reported that they considered PGT in the context of religion, had less interest in PGT.

It is unknown whether the attitudes of FORCE members toward PGT are representative of the majority of BRCA carriers. A cross-sectional study of 1,081 BRCA carriers, 65% of whom were recruited through FORCE and the remainder by the University of Pennsylvania, revealed that a majority of carriers were in favor of offering PGT and prenatal diagnosis to carriers (59% for PGT and 55.5% for prenatal diagnosis).[188] Of those who indicated that their families were not complete, 41% of BRCA carriers reported that their carrier status impacted their decision about future biological children. This study also revealed that 21.5% of unpartnered BRCA carriers felt more pressure to get married.

The U.K. Human Fertilization and Embryology authority has approved the use of PGT for hereditary breast and ovarian cancer. In a sample of 102 women with a BRCA pathogenic variant, most were supportive of PGT but only 38% of the women who had completed their families would consider it for themselves had PGT been available, and only 14% of women who were contemplating a future pregnancy would consider PGT.[189] In a study of 77 individuals undergoing BRCA testing as part of a multicenter cohort study in Spain, 61% of respondents reported they would consider PGT. Factors associated with PGT interest were age 40 years and older and had a prior cancer diagnosis.[190]

In France, couples who obtain authorization from a multidisciplinary prenatal diagnosis team may access PGT free of charge as a benefit of their national health care system. However, no BRCA carriers have been authorized to use PGT. In a national study of 490 unaffected carriers of BRCA pathogenic variants of childbearing age (women aged 18–49 y; men aged 18–69 y), 16% stated that BRCA test results had altered their ongoing plans for childbearing.[191] Upon qualitative analysis of written comments provided by some respondents, the primary impact was related to accelerating the timing of pregnancy, feelings of guilt about possibly passing on the pathogenic variant to offspring, and having future children. In response to a hypothetical scenario in which PGT was readily available, 33% of participants reported that they would undergo PGT. Factors associated with this intention were having no future reproductive plans at the time of the survey, feeling pregnancy termination was an acceptable option in the context of identifying a BRCA pathogenic variant, and having fewer cases of breast and/or ovarian cancer in the family. When presented with questions about expectations about delivery of PGT or prenatal diagnosis (PND) information, 85% of respondents felt it should be provided along with BRCA test results; 45% felt that it should be provided when carriers decide to have children. Respondents stated that they would expect this information to be delivered by cancer geneticists (92%), obstetrician/gynecologists (76%), and general practitioners (48%).

A small (N = 25) qualitative study of women of reproductive age positive for a BRCA pathogenic variant who underwent genetic testing before having children evaluated how their BRCA status influenced their attitudes about reproductive genetic testing (both PGT and PND) and decisions about having children.[192] In this study, the decision to undergo BRCA testing was primarily motivated by the desire to manage one’s personal cancer risk, rather than a desire to inform future reproductive decisions. The perceived severity of HBOC influenced concerns about passing on a BRCA pathogenic variant to children and also influenced willingness to consider PGT or PND and varied based on personal experience. Most did not believe that a known BRCA pathogenic variant was a reason to terminate a pregnancy. As observed in prior studies, knowledge of reproductive options varied; however, there was a tendency among participants to view PGT as more acceptable than PND with regard to termination of pregnancy. Decisions regarding the pros and cons of PGT versus PND with termination of pregnancy were driven primarily by personal preferences and experiences, rather than by morality judgments. For example, women were deterred from PGT based on the need to undergo in vitro fertilization and to take hormones that might increase cancer risk and based on the observed experiences of others who underwent this procedure.

One study has examined these issues among high-risk men recruited from FORCE and Craigslist (a bulletin board website) (N = 228).[193] Similar to the previous studies of women, only 20% of men were aware of PGT before survey participation. In a multivariate analysis, those who selected the “other” option for possible benefits of PGT compared with those who selected from several predetermined options (e.g., having children without genetic variants) and those who considered PGT in the context of religion (as opposed to health and safety) were less likely to report that they would ever consider using PGT.

Cultural/Community Effects

The recognition that BRCA1/BRCA2 pathogenic variants are prevalent, not only in breast/ovarian cancer families but also in some ethnic groups,[194] has led to considerable discussion of the ethical, psychological, and other implications of having one’s ethnicity be a factor in determination of disease predisposition. Concerns that people will think everything is solely determined by genetic factors and the creation of a genetic underclass [195] have been voiced. Questions about the impact on the group of being singled out as having genetic vulnerability to breast cancer have been raised. There is also confusion about who gives or withholds permission for the group to be involved in studies of their genetic identity. These issues challenge traditional views on informed consent as a function of individual autonomy.[196]

A growing literature on the unique factors influencing a variety of cultural subgroups suggests the importance of developing culturally specific genetic counseling and educational approaches.[100,197-201] The inclusion of members within the community of interest (e.g., breast cancer survivors, advocates, and community leaders) may enhance the development of culturally tailored genetic counseling materials.[101] One study showed that participation in any genetic counseling (culturally mediated or standard approaches) reduced perceived risk of developing breast cancer.[202]

Ethical Concerns

The human implications of the ethical issues raised by the advent of genetic testing for breast/ovarian cancer susceptibility are described in case studies,[203] essays,[204,205] and research reports. Issues about rights and responsibilities in families concerning the spread of information about genetic risk promise to be major ethical and legal dilemmas in the coming decades.

Studies have shown that 62% of studied family members were aware of the family history and that 88% of hereditary breast/ovarian cancer family members surveyed have significant concerns about privacy and confidentiality. Expressed concern about cancer in third-degree relatives, or relatives farther removed, was about the same as that for first- or second-degree relatives of the proband.[206] Only half of surveyed FDRs of women with breast or ovarian cancer felt that written permission should be required to disclose BRCA1/BRCA2 test results to a spouse or immediate family member. Attitudes toward testing varied by ethnicity, previous exposure to genetic information, age, optimism, and information style. Altruism is a factor motivating genetic testing in some people.[207] Many professional groups have made recommendations regarding informed consent.[113,207-210] There is some evidence that not all practitioners are aware of or follow these guidelines.[211] Research shows that many BRCA1/BRCA2 genetic testing consent forms do not fulfill recommendations by professional groups about the 11 areas that should be addressed,[212] and they omit highly relevant points of information.[211] In a study of women with a history of breast or ovarian cancer, the interviews yielded that the women reported feeling inadequately prepared for the ethical dilemmas they encountered when imparting genetic information to family members.[213] These data suggest that more preparation about disclosure to family members before testing reduces the emotional burden of disseminating genetic information to family members. Patients and health care providers would benefit from enhanced consideration of the ethical issues of warning family members about hereditary cancer risk. (Refer to the PDQ summaries Cancer Genetics Risk Assessment and Counseling and Cancer Genetics Overview for more information about the ethics of cancer genetics and genetic testing.)

Psychosocial Aspects of Cancer Risk Management for Hereditary Breast and Ovarian Cancer

Decision aids for individuals considering risk management options for hereditary breast and ovarian cancer

There is a small but growing body of literature on the use of decision aids as an adjunct to standard genetic counseling to assist patients in making informed decisions about cancer risk management.[214-218] One study showed that the use of a decision aid consisting of individualized value assessment and cancer risk management information after receiving positive BRCA1/BRCA2 test results was associated with fewer intrusive thoughts and lower levels of depression at the 6-month follow-up in unaffected women. Use of the decision aid did not alter cancer risk management intentions and behaviors. Slightly detrimental effects on well-being and several decision-related outcomes, however, were noted among affected women.[216] Another study compared responses to a tailored decision aid (including a values-clarification exercise) versus a general information pamphlet intended for women making decisions about ovarian cancer risk management. In the short term, the women receiving the tailored decision aid showed a decrease in decisional conflict and increased knowledge compared with women receiving the pamphlet, but no differences in decisional outcomes were found between the two groups. In addition, the decision aid did not appear to alter the participant’s baseline cancer risk management decisions.[215] A multisite randomized trial of 150 unaffected women with BRCA1/BRCA2 pathogenic variants assessed the effect of a decision aid on breast cancer risk management decisions and psychosocial outcomes. At 6-month and 12-month follow-up, women randomly assigned to the decision aid had lower levels of cancer-related distress (P = .01 at 6 months and P = .05 at 12 months) than did the control group.[218] Decisional conflict scores were relatively low at baseline and declined over time in both groups; the scores between the two groups were not statistically different.

Uptake of cancer risk management options

An increasing number of studies have examined uptake and adherence to cancer risk management options among individuals who have undergone genetic counseling and testing for BRCA1 and BRCA2 pathogenic variants. Findings from these studies are reported in Table 15 and Table 16. Outcomes vary across studies and include uptake or adherence to screening (mammography, magnetic resonance imaging [MRI], cancer antigen [CA] 125, transvaginal ultrasound [TVUS]) and selection of RRM and RRSO. Studies generally report outcomes by pathogenic variant carrier or testing status (e.g., positive for pathogenic variants, negative for pathogenic variants, or declined genetic testing). Follow-up time after notification of genetic risk status also varied across studies, ranging from 12 months up to several years.

Findings from these studies suggest that breast screening often improves after notification of BRCA1/BRCA2 pathogenic variant carrier status; nonetheless, screening remains suboptimal. Fewer studies have examined adoption of MRI as a screening modality, probably due to the recent availability of efficacy data. Screening for ovarian cancer varied widely across studies, and also varied based on type of screening test (i.e., CA-125 serum testing vs. TVUS screening). However, ovarian cancer screening does not appear to be widely adopted by carriers of BRCA1/BRCA2 pathogenic variants. Uptake of RRM varied widely across studies and may be influenced by personal factors (such as younger age or having a family history of breast cancer), psychosocial factors (such as a desire for reduction of cancer-related distress), recommendations of the health care provider, and cultural or health care system factors. An individual’s choice to have a bilateral mastectomy also appears to be influenced by pretreatment genetic education and counseling regardless of the genetic test results.[219] Similarly, uptake of RRSO also varied across studies, and may be influenced by similar factors, including older age, personal history of breast cancer, perceived risk of ovarian cancer, cultural factors (i.e., country), and the recommendations of the health care provider.

Table 15. Uptake of Risk-reducing Mastectomy (RRM) and/or Breast Screening Among Carriers of BRCA1 and BRCA2 Pathogenic Variants
Study Citation Study Population Uptake of RRM Uptake of Breast Screening Mammography and/or Breast MRI Length of Follow-up Comments
MRI = magnetic resonance imaging; RRSO = risk-reducing salpingo-oophorectomy.
aSelf-report as data source.
bMedical records as data source.
United States
Botkin et al. (2003) [220] Carriers (n = 37)a Carriers 0% Mammography 24 mo  
– Carriers 57%
Noncarriers (n = 92)a Noncarriers 0% – Noncarriers 49%
– Declined test 20%
Declined testing (n = 15)a   MRI
– Not evaluated
Beattie et al. (2009) [221] Carriers (n = 237)b Carriers 23% Not applicable Mean, 3.7 y Women opting for RRM were younger than 60 y, had a prior diagnosis of breast cancer, and also underwent RRSO
Median time to RRM: 124 days from receiving results
O’Neill et al. (2010) [222] Carriers (n = 146)a Carriers 13% Not applicable 12 mo Intentions at test result disclosure predicted RRM decisions
Schwartz et al. (2012) [223] Carriers (n = 108)a Carriers 37% Mammography Mean, 5.3 y Predictors of RRM were younger age, higher precounseling cancer distress, more recent diagnosis of breast or ovarian cancer, and intact ovaries
– Carriers affected 92%
– Carriers unaffected 82%
Noncarriers (n = 60)a Noncarriers 0% – Noncarriers 66%
– Uninformative affected 89%
MRI
Uninformative (n = 206)a Uninformative 6.8% – Carriers affected 51%
– Carriers unaffected 46%
– Noncarriers 11%
– Uninformative 27%
Garcia et al. (2013) [224] Carriers (n = 250)b Carriers 44% Excluding women post RRM: 41 months; range, 26–66 mo Breast surveillance decreased significantly from y 1–5 of follow-up: Mammography 43% to 7%; MRI 35% to 3%
Mammography:
– Carriers 43%
MRI:
– Carriers 35%
Singh et al. (2013) [225] Carriers (n = 136)b Carriers 42% Not applicable Range, 1–11 y Predictors of RRM were first- or second-degree relative diseased from breast cancer, having had at least one childbirth, and having undergone testing after 2005
International
Phillips et al. (2006) [226] Carriers (n = 70)a Carriers 11% Mammography 3 y  
– Carriers 89%
MRI
– Not evaluated
Metcalfe et al. (2008) [227] Carriers (N = 2,677)a Carriers 18% (unaffected) Mammography 3.9 y; range, 1.5–10.3 y Large differences in uptake of risk management options by country
– Carriers 87%
MRI 1,294 participants had a personal history of breast cancer
– Carriers 31%
Julian-Reynier et al. (2011) [228] Carriers (n = 101)a Carriers 6.9% Mammography 5 y Noncarriers often continued screening
– Carriers 59%
– Noncarriers aged 30–39 y 53%
Noncarriers (n = 145)a Noncarriers 0% MRI
– Carriers 31%
– Noncarriers 4.8%
Table 16. Uptake of Risk-reducing Salpingo-oophorectomy (RRSO) and/or Gynecologic Screening Among Carriers of BRCA1 and BRCA2 Pathogenic Variants
Study Citation Study Population Uptake of RRSO Uptake of Gynecologic Screening Length of Follow-up Comments
CA-125 = cancer antigen 125; RRM = risk-reducing mastectomy; TVUS = transvaginal ultrasound.
aSelf-report as data source.
bMedical records as data source.
cData source not specified.
United States
Scheuer et al. (2002) [229] Carriers (n = 179)a Carriers 50.3% CA-125 Mean, 24.8 mo; range, 1.6–66.0 mo Women undergoing RRSO were older and more likely to have a personal history of breast cancer
– Carriers 67.6%
TVUS
– Carriers 72.9%
Beattie et al. (2009) [221] Carriers (n = 240)b Carriers 51% Not applicable Mean, 3.7 y Women opting for RRSO <60 y had a prior diagnosis of breast cancer and also underwent RRM
Median time to RRSO: 123 days from receiving results
O'Neill et al. (2010) [222] Carriers (n = 146)a Carriers 32% Not applicable 12 mo  
Schwartz et al. (2012) [223] Carriers (n = 100)a Carriers 65% CA-125 Mean, 5.3 y Predictors of RRSO were being ≥40 y and having received a diagnosis of breast cancer more than 10 y ago
Noncarriers (n = 52)a Noncarriers 1.9% – Carriers 56%
– Noncarriers 12%
– Uninformative 33%
Uninformative (n = 203)a Uninformative 13.3% TVUS
– Carriers 42%
– Noncarriers 20%
– Uninformative 26%
Garcia et al. (2013) [224] Carriers (n = 305)b Carriers 74% Excluding women post-RRSO: 41 mo; range, 26–66 mo Ovarian surveillance decreased significantly from years 1–5 of follow-up; CA-125: 47% to 2%; TVUS: 45% to 2.3%
CA-125
– Carriers 47%
TVUS
– Carriers 45%
Mannis et al. (2013) [230] Carriers (n = 201)a Carriers 69.6% CA-125 Median, 3.7 y Predictors of RRSO and screening included being a carrier of a BRCA pathogenic variant, age 40–49 y, having a higher income, ≥2 children, a personal history of breast cancer, and a first-degree relative with ovarian cancer
– 26.3%
TVUS
– 26.3%
Noncarriers (n = 103)a Noncarriers 2.0% Not reported
Uninformative (n = 773)a; 59/773 with a variant of uncertain significance Uninformative 12.3% CA-125
– 10.4%
TVUS
– 6.5%
Singh et al. (2013) [225] Carriers (n = 136)b Carriers 52% Not applicable Range, 1–11 y Predictors of RRSO were first- or second-degree relative with breast cancer, a mother lost to pelvic cancer, having had ≥1 childbirths, age ≥50 y, and having undergone testing after 2005
International
Phillips et al. (2006) [226] Carriers (n = 70)a Carriers 29% CA-125 3 y  
– Carriers 0%
TVUS
– Carriers 67%
Friebel et al. (2007) [231] Carriers (N = 537)c Carriers 55% Not applicable Minimum 6 mo; median 36 mo RRSO greatest in parous women >40 y
Madalinska et al. (2007) [232] Carriers (n = 160)a, b Carriers 74% Carriers 26% 12 mo Women who underwent RRSO had lower education levels, viewed ovarian cancer as incurable, and believed strongly in the benefits of RRSO
Specific method(s) of gynecological screening not reported
Metcalfe et al. (2008) [227] Carriers (N = 2,677)a Carriers 57% Not applicable 3.9 y; range, 1.5–10.3 y Large differences in uptake of risk management options by country
Julian-Reynier et al. (2011) [228] Carriers (n = 101)a Carriers 42.6% TVUS 5 y RRSO uptake increased with age. Having undergone RRSO did not alter breast cancer risk perception. Noncarriers often continued screening
Noncarriers (n = 145)a Noncarriers 2% – Noncarriers 43.2%
Rhiem et al. (2011) [233] Carriers (N = 306)b Carriers 57% Not evaluated Mean, 47.8 mo post-oophorectomy Median age at time of RRSO = 47 y. One occult fallopian tube cancer was detected at the time of RRSO. One peritoneal carcinoma was diagnosed 26 mo post-RRSO
Sidon et al. (2012) [234] Carriers (N = 700)a; 386/700 with personal history of breast cancer BRCA1 carriers: Not evaluated Affected with breast cancer Uptake of RRSO was lower in women >60 y (22% uptake at 5 y). None of the women >70 y had a RRSO performed
– 54.5%
BRCA2 carriers: BRCA1: Mean, 2.29; range, 0.1–11.45 y
– 45.5%
All carriers with no personal history of breast cancer BRCA2: Mean, 1.77; range, 0.1–11.1 y
Not affected with breast cancer
– 54.2%
All carriers with personal history of breast cancer BRCA1: Mean, 1.63; range, 0.1–11.28 y
– 43.2% BRCA2: Mean, 1.75; range, 0.1–8.98 y

On the other hand, many women found to be pathogenic variant carriers express interest in RRM in hopes of minimizing their risk of breast cancer. In one study of a number of unaffected women with no previous risk-reducing surgery who received results of BRCA1 testing after genetic counseling, 17% of carriers (2 of 12) intended to have mastectomies and 33% (4 of 12) intended to have oophorectomies.[235] In a later study of the same population, RRM was considered an important option by 35% of women who tested positive, whereas risk-reducing oophorectomy was considered an important option by 76%. A prospective study assessed the stability of risk management preferences over five time points (pre-BRCA testing to 9 months after results disclosure) among 80 Dutch women with a documented BRCA pathogenic variant. Forty-six participants indicated a preference for screening at baseline. Of 25 women who preferred RRM at baseline, 22 indicated the same preference 9 months after test results disclosure; however, it was not reported how many women actually had RRM.[236]

Initial interest does not always translate into the decision for surgery. Two different studies found low rates of RRM among carriers of pathogenic variants in the year after result disclosure, one showing 3% (1 of 29) of carriers and the other 9% (3 of 34) of carriers having had this surgery.[168,237] Among members from a large BRCA1 kindred, utilization of cancer screening and/or risk-reducing surgeries was assessed at baseline (before disclosure of results), and at 1 year and 2 years after disclosure of BRCA1 test results. Of the 269 men and women who participated, complete data were obtained on 37 female carriers and 92 female noncarriers, all aged 25 years or older. At 2 years after disclosure of test results, none of the women had undergone RRM, although 4 of the 37 carriers (10.8%) said they were considering the procedure. In contrast, of the 26 women who had not had an oophorectomy before baseline, 46% (12 of 26) had obtained an oophorectomy by 2 years after testing. Of those carriers aged 25 to 39 years, 29% (5 of 17) underwent oophorectomy, while 78% (7 of 9) of the carriers aged 40 years and older had this procedure.[220] In a study assessing uptake of risk-reducing surgery 3 months after BRCA result disclosure, 7 of 62 women had undergone RRM and 13 of 62 women had undergone RRSO. Intent to undergo RRSO before testing correlated with procedure uptake. In contrast, intent to undergo RRM did not correlate with uptake. Overall, reasons given for indecision about risk-reducing surgery included complex testing factors such as the significance of family history in the absence of a pathogenic variant, concerns over the surgical procedure, and time and uncertainty regarding early menopause and the use of HRT.[238] In a U.K. study, data were collected during observations of genetic consultations and in semistructured interviews with 41 women after they received genetic counseling.[239] The option of risk-reducing surgery was raised in 29 consultations and discussed in 35 of the postclinic interviews. Fifteen women said they would consider having an oophorectomy in the future, and nine said they would consider having a mastectomy. The implications of undergoing oophorectomy and mastectomy were discussed in postclinic interviews. Risk-reducing surgery was described by the consultands as providing individuals with a means to (a) fulfill their obligations to other family members and (b) reduce risk and contain their fear of cancer. The costs of this form of risk management were described by the respondents as follows:

  • Compromising social obligations.
  • Upsetting the natural balance of the body.
  • Not receiving protection from cancer.
  • Operative and postoperative complications.
  • The onset of menopause.
  • The effects of body image, gender, and personal identity.
  • Potential effects on sexual relationships.[239]

A number of women choose to undergo RRM and RRSO without genetic testing because of the following:

  • Testing is not readily accessible.
  • They do not wish exposure to the psychosocial risks of genetic testing.
  • They do not trust that a negative genetic test result means they are not at increased risk.
  • They find any level of risk, even baseline population risk, unacceptable.[240,241]

Among FDRs of breast cancer patients attending a surveillance clinic, women who expressed an interest in RRM and/or had undergone surgery were found to have significantly more breast cancer biopsies (P < .05) and higher subjective 10-year breast cancer risk estimates (P < .05) than women not interested in RRM. Cancer worry at the time of entry into the clinic was highest among women who subsequently underwent RRM compared with women who expressed interest but had not yet had surgery and women who did not intend to have surgery (P < .001).[242]

BRCA testing, when offered to women newly diagnosed with breast cancer, has been shown to influence surgical decision making in that carriers are more likely to opt for bilateral mastectomy compared with noncarriers.[219,243,244] A study that evaluated predictors of contralateral RRM among 435 breast cancer survivors found that 16% had undergone contralateral RRM (in conjunction with mastectomy of the affected breast) before referral for genetic counseling and BRCA1/BRCA2 genetic testing.[245] Predictors of contralateral RRM before genetic counseling and testing included younger age at breast cancer diagnosis, more time since diagnosis, having at least one affected FDR, and not being employed full-time. In the year after disclosure of test results, 18% of women who tested positive for a BRCA1/BRCA2 pathogenic variant and 2% of those whose test results were uninformative underwent contralateral RRM. Predictors of contralateral RRM after genetic testing included younger age at breast cancer diagnosis, higher cancer-specific distress before genetic counseling, and having a positive BRCA1/BRCA2 test result. In this study, contralateral RRM was not associated with distress at 1 year after disclosure of genetic test results. A retrospective chart review evaluated uptake of bilateral mastectomies in 110 women who underwent BRCA1/BRCA2 genetic testing before making surgical decisions about the treatment of newly diagnosed breast cancer. Carriers of BRCA pathogenic variants were more likely to undergo bilateral mastectomies than were women in whom no variant was detected (83% vs. 37%; P = .046).[246] The only predictor of contralateral RRM in women without a pathogenic variant was being married (P = .03). Age, race, parity, disease stage and biomarkers, increased mammographic breast density, and breast MRI did not influence contralateral RRM decisions at the time of primary surgical treatment.

A study conducted from 2006 to 2014 in 11 U.S. academic and community centers of 897 women, aged 40 years and younger at breast cancer diagnosis, found that rates of BRCA genetic testing have increased over time.[244] Within 1 year after diagnosis, 87% of the sample underwent BRCA testing. The rate increased from 77% of newly diagnosed women tested in 2006 to 95% of women tested in 2013. Among women who tested positive for a pathogenic BRCA variant and stated that testing affected their surgery decisions (n = 88), 86% underwent bilateral mastectomy compared with 51% of noncarriers (P < .001). Among untested women, about one-third reported that they were not told by a health care provider that they were candidates for BRCA testing; yet, according to national guidelines, all were eligible for testing solely on the basis of their age at diagnosis.

Dutch women (N = 114) who had undergone unilateral or bilateral RRM with breast reconstruction between 1994 and 2002 were retrospectively surveyed to determine their satisfaction with the procedure.[247] Sixty-eight percent were either unaffected carriers of BRCA pathogenic variants or at a 50% risk of having a BRCA pathogenic variant in their family. Sixty percent of respondents indicated that they were satisfied with the procedure, 95% would opt for RRM again, and 80% would opt for the same reconstruction procedure. Less than half reported some perioperative or postoperative complications, ongoing physical complaints, or some physical limitations. Twenty-nine percent reported altered feelings of femininity after the procedure, 44% reported adverse changes in their sexual relationships, and 35% indicated that they believed their partners experienced adverse changes in their sexual relationship. Ten percent of women, however, reported positive changes in their sexual relationship after the procedure. Compared with patients who indicated satisfaction with this procedure, nonsatisfied patients were more likely to feel less informed about the procedure and its consequences, report more complications and physical complaints, feel that their breasts did not belong to their body, and indicate that they would not opt for reconstruction again. Those who reported a negative effect on their sexual relationship were more likely to:

  • Feel less informed.
  • Experience more physical complaints and limitations.
  • Express that their breasts did not feel like their own.
  • Be disinclined to opt for reconstruction again.
  • State that the surgery had not met their expectations.
  • Experience altered feelings of femininity and perceived adverse changes in their partner’s view of their femininity and their sexual relationship.

Ninety Swedish women who had undergone RRM between 1997 and 2005 were surveyed before surgery, 6 months after surgery, and 1 year after surgery to evaluate changes in health-related quality of life, depression, anxiety, sexuality, and body image. There were no significant changes in health-related quality of life or depression at the three time points; anxiety decreased over time (P = .0004). More than 80% of women reported having an intimate relationship at all three time points. Women who reported being sexually active were asked to respond to questions about sexual pleasure, discomfort, habit, and frequency of activity. There were no statistically significant differences related to frequency, habit, or discomfort. However, pleasure significantly decreased between baseline and 1 year after surgery (P = .005). At 1 year after surgery, 48% of women reported feeling less attractive, 48% reported feeling self-conscious, and 44% reported dissatisfaction with surgical scars.[248]

Discussion of risk-reducing surgical options may not consistently occur during pretest genetic counseling. In one multi-institutional study, only one-half of genetics specialists discussed RRM and RRSO in consultations with women from high-risk breast cancer families,[249,250] despite the fact that discussion of surgical options was significantly associated with meeting consultands' expectations, and that such information was not associated with increased anxiety.[251]

Given the increased risk of ovarian cancer faced by women with a BRCA1 or BRCA2 pathogenic variant, those who do receive information about RRSO show wide variations in surgery uptake (27%–72%).[9,124,229,232,252,253] A study showed that clinical factors related to choosing RRSO versus surveillance alone are older age, parity of one or more, and a prior breast cancer diagnosis.[254] In this study, the choice of RRSO was not related to family history of breast or ovarian cancer. Hysterectomy was presented as an option during genetic counseling, and 80% of women who underwent RRSO also elected to have a hysterectomy.

Cancer screening and risk-reducing behaviors

Data are now emerging regarding uptake and adherence to cancer risk management recommendations such as screening and risk-reducing interventions. Cancer screening adherence and risk-reduction behaviors as defined by the National Comprehensive Cancer Network Guidelines were assessed in a cross-sectional study of 214 women with a personal history (n = 134) or family history (n = 80) of breast or ovarian cancer. Among unaffected women older than 40 years, 10% had not had a mammogram or clinical breast examination (CBE) in the previous year and 46% did not practice breast self-examination (BSE). Among women previously affected with breast or ovarian cancer, 21% had not had a mammogram, 32% had not had a CBE, and 39% did not practice BSE.[255]

Three hundred and twelve women who were counseled and tested for BRCA pathogenic variants between 1997 and 2005 responded to a survey regarding their perception of genetic testing for hereditary breast and ovarian cancer. The survey included questions on risk reduction options, including screening and risk-reducing surgeries. Two hundred and seventeen (70%) of the women had been diagnosed with breast cancer, and 86 (28%) tested positive for a pathogenic variant in either the BRCA1 or BRCA2 gene. None of the BRCA-positive women agreed that mammograms are difficult procedures because of the discomfort, while 11 (5.4%) of the BRCA-negative women did agree with this statement. Both groups (BRCA-positive and BRCA-negative) agreed that risk-reducing surgeries provide the best means for lowering cancer risk and worry, and most patients in both groups expressed the belief that risk-reducing mastectomy is not too drastic, too scary, or too disfiguring.[256]

A prospective study from the United Kingdom examined the psychological impact of mammographic screening in 1,286 women aged 35 to 49 years who have a family history of breast cancer and were participants in a multicenter screening program. Mammographic abnormalities that required additional evaluation were detected in 112 women. These women, however, did not show a statistically significant increase in cancer worry or negative psychological consequences as a result of these findings. The 1,174 women who had no mammographic abnormality detected experienced a decrease in cancer worry and a decrease in negative psychological consequences compared with baseline after receipt of their results. At 6 months, the entire cohort had experienced a decrease in measures of cancer worry and psychological consequences of breast screening.[257]

A qualitative study explored health care professionals’ views regarding the provision of information about health protective behaviors (e.g., exercise and diet). Seven medical specialists and ten genetic counselors were interviewed during a focus group or individually. The study reported wide variation in the content and extent of information provided about health-protective behaviors and in general, participants did not consider it their role to promote such behaviors in the context of a genetic counseling session. There was agreement, however, about the need to form consensus about provision of such information both within and across risk assessment clinics.[258]

Not all studies specify whether screening uptake rates fall within recommended guidelines for the targeted population or the specific clinical scenario, nor do they report on other variables that may influence cancer screening recommendations. For example, women who have a history of atypical ductal hyperplasia would be advised to follow screening recommendations that may differ from those of the general population.

Psychosocial Outcome Studies

Risk-reducing mastectomy

A prospective study conducted in the Netherlands found that among 26 carriers of BRCA1/BRCA2 pathogenic variants, the 14 women who chose mastectomy had higher distress both before test result disclosure and 6 and 12 months later, compared with the 12 carriers who chose surveillance and compared with 53 women negative for a pathogenic variant. Overall, however, anxiety declined in women undergoing risk-reducing mastectomy (RRM); at 1 year, their anxiety scores were closer to those of women choosing surveillance and to the scores of women negative for a pathogenic variant.[259] Interestingly, women opting for RRM had lower pretest satisfaction with their breasts and general body image than carriers who opted for surveillance or noncarriers of BRCA1/BRCA2 pathogenic variants. Of the women who had an RRM, all but one did not regret the decision at 1 year posttest disclosure, but many had difficulties with body image, sexual interest and functioning, and self-esteem. The perception that doctors had inadequately informed them about the consequences of RRM was associated with regret.[259] At the 5-year follow-up, women who had undergone RRM had less favorable body image and changes in sexual relationships, but also had a significant reduction in the fear of developing cancer.[260] In a study of 78 women who underwent risk-reducing surgery (including BRCA1/BRCA2 carriers and women who were from high-risk families with no detectable BRCA1/BRCA2 pathogenic variant), cancer-specific and general distress were assessed 2 weeks before surgery and at 6 and 12 months postsurgery.[261] The sample included women who had RRM and RRSO alone and women who had both surgeries. There was no observable increase in distress over the 1-year period.

Mixed psychosocial outcomes were reported in a follow-up study (mean 14 years) of 609 women who received RRM at the Mayo Clinic. Seventy percent were satisfied with RRM, 11% were neutral, and 19% were dissatisfied. Eighteen percent believed that if they had the choice to make again, they probably or definitely would not have an RRM. About three-quarters said their worry about cancer was diminished by surgery. One-half reported no change in their satisfaction with body image; 16% reported improved body image after surgery. Thirty-six percent said they were dissatisfied with their body image after RRM. About one-quarter of the women reported adverse impact of RRM on their sexual relationships and sense of femininity, and 18% had diminished self-esteem. Factors most strongly associated with satisfaction with RRM were postsurgical satisfaction with appearance, reduced stress, no reconstruction or lack of problems with implants, and no change or improvement in sexual relationships. Women who cited physician advice as the primary reason for choosing RRM tended to be dissatisfied after RRM.[262]

A study of 60 healthy women who underwent RRM measured levels of satisfaction, body image, sexual functioning, intrusion and avoidance, and current psychological status at a mean of 4 years and 4 months postsurgery. Of this group, 76.7% had either a strong family history (21.7%) or carried a BRCA1 or BRCA2 pathogenic variant (55%). Overall, 97% of the women surveyed were either satisfied (17%) or extremely satisfied (80%) with their decision to have RRM, and all but one participant would recommend this procedure to other women. Most women (66.7%) reported that surgery had no impact on their sexual life, although 31.7% reported a worsening sexual life, and 76.6% reported either no change in body image or an improvement in body image, regardless of whether reconstruction was performed. Worsening self-image was reported by 23.3% of women after surgery. Women’s mean distress levels after surgery were only slightly above normal levels, although those women who continued to perceive their postsurgery breast cancer risk as high had higher mean levels of global and cancer-related distress than those who perceived their risk as low. Additionally, carriers of BRCA1 and BRCA2 pathogenic variants and women with a strong family history of breast and/or ovarian cancer had higher mean levels of cancer-related distress than women with a limited family history.[263]

Very little is known about how the results of genetic testing affect treatment decisions at the time of cancer diagnosis. Two studies explored genetic counseling and BRCA1/BRCA2 genetic testing at the time of breast cancer diagnosis.[24,219] One of these studies found that genetic testing at the time of diagnosis significantly altered surgical decision making, with more pathogenic variant carriers than noncarriers opting for bilateral mastectomy. Bilateral RRM was chosen by 48% of women with a known pathogenic variant [219] and by 100% of women with a known pathogenic variant in a smaller series [24] of women undergoing testing at the time of diagnosis. Of women in whom no pathogenic variant was found, 24% also opted for bilateral RRM. Four percent of the test decliners also underwent bilateral RRM. Among carriers of pathogenic variants, predictors of bilateral RRM included whether patients reported that their physicians had recommended BRCA1/BRCA2 testing and bilateral RRM before testing, and whether they received a positive test result.[219] Data are lacking on quality-of-life outcomes for women who undergo RRM after genetic testing that is performed at the time of diagnosis.

A prospective study from the Netherlands evaluated long-term psychological outcomes of offering women with breast cancer genetic counseling and, if indicated, genetic testing at the onset of breast radiation for treatment of their primary breast cancer. Of those who were approached for counseling, some underwent genetic testing and chose to receive their result (n = 58), some were approached but did not fulfill referral criteria (n = 118), and some declined the option of counseling/testing (n = 44). Another subset of women undergoing radiation therapy was not approached for counseling (n = 182) but was followed using the same measures. Psychological distress was measured at baseline and at 4, 11, 27, and 43 weeks after initial consultation for radiation therapy. No differences were detected in general anxiety, depression or breast cancer–specific distress across all four groups.[264]

A retrospective questionnaire study of 583 women with a personal and family history of breast cancer and who underwent contralateral RRM between 1960 and 1993 measured overall satisfaction after mastectomy and factors influencing satisfaction and dissatisfaction with this procedure.[265] The mean time of follow-up was 10.3 years after risk-reducing surgery. Overall, 83% of all participants stated they were satisfied or very satisfied, 8% were neutral, and 9% were dissatisfied with contralateral RRM. Most women also reported favorable effects or no change in their self-esteem, level of stress, and emotional stability after surgery (88%, 83%, and 88%, respectively). Despite the high levels of overall satisfaction, 33% reported negative body image, 26% reported a reduced sense of femininity, and 23% reported a negative effect on sexual relationships. The type of surgical procedure also affected levels of satisfaction. The authors attributed this difference to the high rate of unanticipated reoperations in the group of women having subcutaneous mastectomy (43%) versus the group having simple mastectomy (15%) (P < .0001). Limitations to this study are mostly related to the time period during which participants had their surgery (i.e., availability of surgical reconstructive option).[265,266] None of these women had genetic testing for pathogenic variants in the BRCA1/BRCA2 genes. Nevertheless, this study shows that while most women in this group were satisfied with contralateral RRM, all women reported at least one adverse outcome.

A retrospective survey of 137 BRCA carriers examined the psychosocial impact of preserving the nipple-areolar complex (NAC) in women with bilateral RRM.[267] The study found that body image and sexual well-being differed significantly based on the type of RRM the women underwent. Women with NAC preservation were more satisfied with their breasts (72% vs. 61%), were more satisfied with the surgical outcome (85% vs. 74%) and had greater sexual well-being (68% vs. 52%) than women without NAC preservation. No differences in cancer-related distress, anxiety, depression, or risk perceptions were observed between the two groups. Oncologic outcomes of nipple-sparing mastectomy in BRCA carriers have not been inferior to RRM without NAC preservation.[268] (Refer to the RRM section of this summary for more information.)

Another study compared long-term quality-of-life outcomes in 195 women after bilateral RRM performed between 1979 and 1999 versus 117 women at high risk of breast cancer opting for screening. No statistically significant differences were detected between the groups for psychosocial outcomes. Eighty-four percent of those opting for surgery reported satisfaction with their decision. Sixty-one percent of women from both the surgery and screening groups reported being very much or quite a bit contented with their quality of life.[269]

In a study of psychosocial outcomes associated with RRM and immediate reconstruction, 61 high-risk women (27 carriers of pathogenic variants, others with high-risk family history), 31 of whom had a prior history of breast cancer, were evaluated on average 3 to 4 years after surgery.[270] The study utilized questions designed to elicit yes versus no responses and found that the surgery was well-tolerated with 83% of participants reporting that the results of their reconstructive surgery were as they expected, 90% reporting that they had received adequate preoperative information, none reporting that they regretted the surgery, and all reporting that they would choose the same route if they had to do it again. Satisfaction with the results ranged from 74% satisfied with the shape of their breasts to 89% satisfied with the appearance of the scarring. Comparison of this group to normative samples on quality-of-life indicators (Short Form 36 Health Survey Questionnaire [SF-36]; Hospital Anxiety and Depression Scale questionnaire scores) indicated no reductions in quality of life in these women.

A qualitative study examining material on the FORCE website posted by 21 high-risk women (BRCA1/BRCA2 positive) undergoing RRM showed that these women anticipated and received negative reactions from friends and family regarding the surgery, and that they managed disclosure in ways to maintain emotional support and self-protection for their decision. Many of the women expressed a relief from intrusive breast cancer thoughts and worry, and were satisfied with the cosmetic result of their surgery.[271]

In contrast, another study examined long-term psychosocial outcomes in 684 women who had had bilateral or contralateral RRM on average 9 years before assessment.[272] A majority of women (59%) also had reconstructive surgery. Interestingly, based on a Likert scale, 85% of women reported that they were satisfied or very satisfied with their decision to have an RRM. However, in qualitative interviews, a large number of women went on to describe dissatisfaction or negative psychosocial outcomes associated with surgery. The authors coded the responses as negative when women reported still being anxious about their breast cancer risk and/or reported negative feelings about their body image, pain, and sexuality. Seventy-nine percent of the women providing negative comments and 84% of those making mixed comments (mixture of satisfaction and dissatisfaction) responded that they were either satisfied or very satisfied with their decision. Twice as many women with bilateral mastectomy made negative and mixed comments than did women with contralateral mastectomy. The areas of most concern were body image, problems with breast implants, pain after surgery, and sexuality. The authors proposed that those who had undergone contralateral procedures had already been treated for cancer, while those who had undergone bilateral procedures had not been treated previously, and this may partially account for the differences in satisfaction between the two groups. These findings suggest that women's satisfaction with RRM may be tempered by their complex reactions over time.

In a qualitative study of 108 women who underwent or were considering RRM, more than half of those who had RRM felt that presurgical consultation with a psychologist was advisable; nearly two-thirds thought that postsurgical consultation was also appropriate. All of the women who were considering RRM believed that psychological consultation before surgery would facilitate decision-making.[273]

Risk-reducing salpingo-oophorectomy

A retrospective self-administered survey of 40 women aged 35 to 74 years at time of RRSO (57.5% were younger than 50 y), who had undergone the procedure through the Ontario Ministry of Health due to a family history of ovarian cancer, found that RRSO resulted in a significant reduction in perceived ovarian cancer risk. Fifty-seven percent identified a decrease in perceived risk as a benefit of RRSO (35% did not comment on RRSO benefits) and 49% reported that they would repeat RRSO to decrease cancer risk. The overall quality-of-life scores were consistent with those published for women who are menopausal or participating in hormone studies.[274] Quality of life in 59 women who underwent RRSO was assessed at 24 months postprocedure.[275] Overall quality of life was similar to the general population and breast cancer survivors, with approximately 20% reporting depression. The 30% of subjects reporting vaginal dryness and dyspareunia were more likely to report dissatisfaction with the procedure.

A Canadian prospective study examined the impact of RRSO on menopausal symptoms and sexual functioning before surgery and then 1 year later in a sample of 114 women with known BRCA1/BRCA2 pathogenic variants.[276] Satisfaction with the decision to undergo RRSO was high regardless of symptoms reported. Those who were premenopausal at the time of surgery (n = 75) experienced a worsening of symptoms and a decline in sexual functioning. HRT addressed vaginal dryness and dyspareunia but not declines in sexual pleasure. HRT also resulted in fewer moderate to severe hot flashes.

Additional work reported by this group found that the majority of the 127 women who had undergone RRSO 1 year previously (75 with BRCA1 pathogenic variants; 52 with BRCA2 pathogenic variants) felt that RRSO reduced their risk of both breast and ovarian cancer.[277] There was a wide range of risk perceptions for ovarian cancer noted in the group. Twenty percent of carriers of BRCA1 and BRCA2 pathogenic variants thought that their risk of ovarian cancer was completely eliminated; others had an inflated perception of their ovarian cancer risk both before and after surgery. A small group of these women were further surveyed at about 3 years postsurgery, and their risk perceptions did not change significantly during this extended time period. These findings suggest that important misperceptions about ovarian cancer risk may persist after RRSO. Additional genetic education and counseling may be warranted.

A larger study assessed quality of life in women at high risk of ovarian cancer who opted for periodic gynecologic screening (GS) versus those who underwent RRSO. Eight hundred forty-six high-risk women, 44% of whom underwent RRSO and 56% of whom chose GS, completed questionnaires evaluating quality of life, cancer-specific distress, endocrine symptoms, and sexual functioning.[278] Women in the RRSO group were a mean of 2.8 ±1.9 years from surgery and women in the GS group were a mean of 4.3 years from their first visit to a gynecologist for high-risk management. No statistical difference in overall quality of life was detected between the RRSO and GS groups. When compared with the GS group, women who underwent RRSO had poorer sexual functioning and more endocrine symptoms such as vaginal dryness, dyspareunia, and hot flashes. Women who underwent RRSO experienced lower levels of breast and ovarian cancer distress and had a more favorable perception of cancer risk.

Women (N = 182) at risk of hereditary breast and ovarian cancer referred for genetic counseling were surveyed concerning their satisfaction with their choice of either RRSO or periodic screening (PS) (biannual pelvic examination with TVUS and CA-125 determination) to manage their ovarian cancer risk.[279] Overall satisfaction with both options was extremely high, but highest among those who chose RRSO over PS. There were no other demographic or clinical factors that distinguished satisfaction level. There was higher decisional ambivalence among those who chose PS.

A retrospective study assessed 98 carriers of BRCA pathogenic variants who underwent RRSO about their preoperative counseling regarding symptoms to expect after surgery.[280] The mean age at RRSO was 45.5 years (range, 32–63 y). Eighty-five percent pursued RRSO after learning that they harbored a BRCA pathogenic variant, and 48.0% were premenopausal at the time of surgery. Participants reported ‘‘frequent’’ or ‘‘very frequent’’ postsurgical symptoms of vaginal dryness (52.1%), changes in interest in sex (50.0%), sleep disturbances (46.7%), changes in sex life (43.9%), and hot flashes (42.9%). Only vaginal dryness and hot flashes were commonly addressed preoperatively. While 96% would have the surgery again, participants reported that the discussion of the impact of surgery on their sex life (59.2%), risk of coronary heart disease (57.1%), and the availability of sex counseling (57.1%) would have been helpful.

Behavioral Outcomes

A study [281] of screening behaviors of 216 self-referred, high-risk (>10% risk of carrying a BRCA1/BRCA2 pathogenic variant) women who are members of hereditary breast cancer families found a range of screening practices. Even the presence of known pathogenic variants in their families was not associated with good adherence to recommended screening practices. Sixty-nine percent of women aged 50 to 64 years and 83% of women aged 40 to 49 years had had a screening mammogram in the previous year. Twenty percent of participants had ever had a CA-125 test and 31% had ever had a pelvic ultrasound or TVUS. Further analysis of this study population [281] looking specifically at 107 women with informative BRCA test results found good use of breast cancer screening, though the uptake rate in younger carriers is lower. The reason for the lower uptake rate was not explored in this study.[282] One survey of screening behaviors among women at increased risk of breast/ovarian cancer identified physician recommendations as a significant factor in adherence to screening.[283]

While motivations cited for pursuing genetic testing often include the expectation that carriers of pathogenic variants will be more compliant with breast and/or ovarian screening recommendations,[281,284-286] limited data exist about whether participants in genetic testing alter their screening behaviors over time and about other variables that may influence those behaviors, such as insurance coverage and physician recommendations or attitudes. The impact of cancer genetic counseling on screening behaviors was assessed in a U.K. study of 293 women followed for 12 months postcounseling at four cancer genetics clinics.[287] BSE, CBE, and mammography were significantly increased after counseling; however, gaps in adherence to recommendations were noted: 38% of women aged 35 to 49 years had not had a mammogram by 12 months postcounseling. BSE was not done by most women at the recommended time and frequency.

This is a critical issue not only for women testing positive, but also for adherence to screening for those testing negative and those who have received indeterminate results or choose not to receive their results. It is possible that adherence actually diminishes with a decrease in the perceived risk that may result from a negative genetic test result.

In addition, while there is still some question regarding the link between cancer-related worry and breast cancer screening behavior, accumulating evidence appears to support a linear rather than a curvilinear relationship. That is, for some time, the data were not consistent; some data supported the hypothesis that mild-to-moderate worry may increase adherence, while excessive worry may actually decrease the utilization of recommended screening practices. Other reports support the notion that a linear relationship is more likely; that is, more worry increases adherence to screening recommendations. Few studies, however, have followed women to assess their health behaviors after genetic testing. Thus, a negative test result leading to decreased worry could theoretically result in decreased screening adherence. A large study found that patient compliance with screening practices was not related to general or screening-specific anxiety—with the exception of BSE, for which compliance is negatively associated with procedure-specific anxiety.[77] Further research designed to clarify this potential concern would highlight the need for comprehensive genetic counseling to discuss the need for follow-up screening.

Further complicating this area of research are issues such as the baseline rate of mammography adherence among women older than 40 or 50 years before genetic testing. More specifically, the ability to note a significant difference in adherence on this measure may be affected by the high adherence rate to this screening behavior before genetic testing by women undergoing such testing. It may be easier to find significant changes in mammography use among women with a family history of breast cancer who test positive. Finally, adherence over time will likely be affected by how women undergoing genetic testing and their caregivers perceive the efficacy of many of the screening options in question, such as mammography for younger women, BSE, and ovarian cancer screening (periodic vaginal ultrasound and serum CA-125 measurements), along with the value of preventive interventions.

The issue of screening decision-making and adherence among women undergoing genetic testing for breast and ovarian cancer is the subject of several ongoing trials, and an area of much needed ongoing study.

References
  1. Ropka ME, Wenzel J, Phillips EK, et al.: Uptake rates for breast cancer genetic testing: a systematic review. Cancer Epidemiol Biomarkers Prev 15 (5): 840-55, 2006. [PUBMED Abstract]
  2. Schwartz MD, Lerman C, Brogan B, et al.: Utilization of BRCA1/BRCA2 mutation testing in newly diagnosed breast cancer patients. Cancer Epidemiol Biomarkers Prev 14 (4): 1003-7, 2005. [PUBMED Abstract]
  3. Kieran S, Loescher LJ, Lim KH: The role of financial factors in acceptance of clinical BRCA genetic testing. Genet Test 11 (1): 101-10, 2007. [PUBMED Abstract]
  4. Susswein LR, Skrzynia C, Lange LA, et al.: Increased uptake of BRCA1/2 genetic testing among African American women with a recent diagnosis of breast cancer. J Clin Oncol 26 (1): 32-6, 2008. [PUBMED Abstract]
  5. Olaya W, Esquivel P, Wong JH, et al.: Disparities in BRCA testing: when insurance coverage is not a barrier. Am J Surg 198 (4): 562-5, 2009. [PUBMED Abstract]
  6. Levy DE, Byfield SD, Comstock CB, et al.: Underutilization of BRCA1/2 testing to guide breast cancer treatment: black and Hispanic women particularly at risk. Genet Med 13 (4): 349-55, 2011. [PUBMED Abstract]
  7. Landsbergen K, Verhaak C, Kraaimaat F, et al.: Genetic uptake in BRCA-mutation families is related to emotional and behavioral communication characteristics of index patients. Fam Cancer 4 (2): 115-9, 2005. [PUBMED Abstract]
  8. Denayer L, Boogaerts A, Philippe K, et al.: BRCA1/2 predictive testing and gender: uptake, motivation and psychological characteristics. Genet Couns 20 (4): 293-305, 2009. [PUBMED Abstract]
  9. Meijers-Heijboer EJ, Verhoog LC, Brekelmans CT, et al.: Presymptomatic DNA testing and prophylactic surgery in families with a BRCA1 or BRCA2 mutation. Lancet 355 (9220): 2015-20, 2000. [PUBMED Abstract]
  10. Wakefield CE, Ratnayake P, Meiser B, et al.: "For all my family's sake, I should go and find out": an Australian report on genetic counseling and testing uptake in individuals at high risk of breast and/or ovarian cancer. Genet Test Mol Biomarkers 15 (6): 379-85, 2011. [PUBMED Abstract]
  11. Schlich-Bakker KJ, ten Kroode HF, Wárlám-Rodenhuis CC, et al.: Barriers to participating in genetic counseling and BRCA testing during primary treatment for breast cancer. Genet Med 9 (11): 766-77, 2007. [PUBMED Abstract]
  12. Vadaparampil ST, McIntyre J, Quinn GP: Awareness, perceptions, and provider recommendation related to genetic testing for hereditary breast cancer risk among at-risk Hispanic women: similarities and variations by sub-ethnicity. J Genet Couns 19 (6): 618-29, 2010. [PUBMED Abstract]
  13. Meiser B: Psychological impact of genetic testing for cancer susceptibility: an update of the literature. Psychooncology 14 (12): 1060-74, 2005. [PUBMED Abstract]
  14. Pasacreta JV: Psychosocial issues associated with genetic testing for breast and ovarian cancer risk: an integrative review. Cancer Invest 21 (4): 588-623, 2003. [PUBMED Abstract]
  15. Simon MS, Petrucelli N: Hereditary breast and ovarian cancer syndrome : the impact of race on uptake of genetic counseling and testing. Methods Mol Biol 471: 487-500, 2009. [PUBMED Abstract]
  16. Hann KEJ, Freeman M, Fraser L, et al.: Awareness, knowledge, perceptions, and attitudes towards genetic testing for cancer risk among ethnic minority groups: a systematic review. BMC Public Health 17 (1): 503, 2017. [PUBMED Abstract]
  17. Meiser B, Gaff C, Julian-Reynier C, et al.: International perspectives on genetic counseling and testing for breast cancer risk. Breast Dis 27: 109-25, 2006-2007. [PUBMED Abstract]
  18. Armstrong K, Stopfer J, Calzone K, et al.: What does my doctor think? Preferences for knowing the doctor's opinion among women considering clinical testing for BRCA1/2 mutations. Genet Test 6 (2): 115-8, 2002 Summer. [PUBMED Abstract]
  19. McCuaig JM, Greenwood CM, Shuman C, et al.: Breast and ovarian cancer: the forgotten paternal contribution. J Genet Couns 20 (5): 442-9, 2011. [PUBMED Abstract]
  20. Burke W, Culver J, Pinsky L, et al.: Genetic assessment of breast cancer risk in primary care practice. Am J Med Genet A 149A (3): 349-56, 2009. [PUBMED Abstract]
  21. Mouchawar J, Klein CE, Mullineaux L: Colorado family physicians' knowledge of hereditary breast cancer and related practice. J Cancer Educ 16 (1): 33-7, 2001. [PUBMED Abstract]
  22. Yong MC, Zhou XJ, Lee SC: The importance of paternal family history in hereditary breast cancer is underappreciated by health care professionals. Oncology 64 (3): 220-6, 2003. [PUBMED Abstract]
  23. Bellcross CA, Leadbetter S, Alford SH, et al.: Prevalence and healthcare actions of women in a large health system with a family history meeting the 2005 USPSTF recommendation for BRCA genetic counseling referral. Cancer Epidemiol Biomarkers Prev 22 (4): 728-35, 2013. [PUBMED Abstract]
  24. Weitzel JN, McCaffrey SM, Nedelcu R, et al.: Effect of genetic cancer risk assessment on surgical decisions at breast cancer diagnosis. Arch Surg 138 (12): 1323-8; discussion 1329, 2003. [PUBMED Abstract]
  25. Kurian AW, Griffith KA, Hamilton AS, et al.: Genetic Testing and Counseling Among Patients With Newly Diagnosed Breast Cancer . JAMA 317 (5): 531-534, 2017. [PUBMED Abstract]
  26. Childers CP, Childers KK, Maggard-Gibbons M, et al.: National Estimates of Genetic Testing in Women With a History of Breast or Ovarian Cancer. J Clin Oncol 35 (34): 3800-3806, 2017. [PUBMED Abstract]
  27. Pal T, Bonner D, Kim J, et al.: Early onset breast cancer in a registry-based sample of African-american women: BRCA mutation prevalence, and other personal and system-level clinical characteristics. Breast J 19 (2): 189-92, 2013 Mar-Apr. [PUBMED Abstract]
  28. Weldon CB, Trosman JR, Gradishar WJ, et al.: Barriers to the use of personalized medicine in breast cancer. J Oncol Pract 8 (4): e24-31, 2012. [PUBMED Abstract]
  29. Hafertepen L, Pastorino A, Morman N, et al.: Barriers to genetic testing in newly diagnosed breast cancer patients: Do surgeons limit testing? Am J Surg 214 (1): 105-110, 2017. [PUBMED Abstract]
  30. Katz SJ, Ward KC, Hamilton AS, et al.: Gaps in Receipt of Clinically Indicated Genetic Counseling After Diagnosis of Breast Cancer. J Clin Oncol 36 (12): 1218-1224, 2018. [PUBMED Abstract]
  31. Schwartz MD, Peshkin BN, Isaacs C, et al.: Randomized trial of proactive rapid genetic counseling versus usual care for newly diagnosed breast cancer patients. Breast Cancer Res Treat 170 (3): 517-524, 2018. [PUBMED Abstract]
  32. Lerman C, Hughes C, Benkendorf JL, et al.: Racial differences in testing motivation and psychological distress following pretest education for BRCA1 gene testing. Cancer Epidemiol Biomarkers Prev 8 (4 Pt 2): 361-7, 1999. [PUBMED Abstract]
  33. Armstrong K, Micco E, Carney A, et al.: Racial differences in the use of BRCA1/2 testing among women with a family history of breast or ovarian cancer. JAMA 293 (14): 1729-36, 2005. [PUBMED Abstract]
  34. Kinney AY, Simonsen SE, Baty BJ, et al.: Acceptance of genetic testing for hereditary breast ovarian cancer among study enrollees from an African American kindred. Am J Med Genet A 140 (8): 813-26, 2006. [PUBMED Abstract]
  35. Halbert CH, Kessler L, Stopfer JE, et al.: Low rates of acceptance of BRCA1 and BRCA2 test results among African American women at increased risk for hereditary breast-ovarian cancer. Genet Med 8 (9): 576-82, 2006. [PUBMED Abstract]
  36. Thompson HS, Valdimarsdottir HB, Duteau-Buck C, et al.: Psychosocial predictors of BRCA counseling and testing decisions among urban African-American women. Cancer Epidemiol Biomarkers Prev 11 (12): 1579-85, 2002. [PUBMED Abstract]
  37. Kinney AY, Gammon A, Coxworth J, et al.: Exploring attitudes, beliefs, and communication preferences of Latino community members regarding BRCA1/2 mutation testing and preventive strategies. Genet Med 12 (2): 105-15, 2010. [PUBMED Abstract]
  38. Sussner KM, Edwards T, Villagra C, et al.: BRCA genetic counseling among at-risk Latinas in New York City: new beliefs shape new generation. J Genet Couns 24 (1): 134-48, 2015. [PUBMED Abstract]
  39. Zimmerman RK, Tabbarah M, Nowalk MP, et al.: Racial differences in beliefs about genetic screening among patients at inner-city neighborhood health centers. J Natl Med Assoc 98 (3): 370-7, 2006. [PUBMED Abstract]
  40. MacNew HG, Rudolph R, Brower ST, et al.: Assessing the knowledge and attitudes regarding genetic testing for breast cancer risk in our region of southeastern Georgia. Breast J 16 (2): 189-92, 2010. [PUBMED Abstract]
  41. Sussner KM, Thompson HS, Jandorf L, et al.: The influence of acculturation and breast cancer-specific distress on perceived barriers to genetic testing for breast cancer among women of African descent. Psychooncology 18 (9): 945-55, 2009. [PUBMED Abstract]
  42. Edwards TA, Thompson HS, Kwate NO, et al.: Association between temporal orientation and attitudes about BRCA1/2 testing among women of African descent with family histories of breast cancer. Patient Educ Couns 72 (2): 276-82, 2008. [PUBMED Abstract]
  43. McCarthy AM, Bristol M, Domchek SM, et al.: Health Care Segregation, Physician Recommendation, and Racial Disparities in BRCA1/2 Testing Among Women With Breast Cancer. J Clin Oncol 34 (22): 2610-8, 2016. [PUBMED Abstract]
  44. Hurtado-de-Mendoza A, Jackson MC, Anderson L, et al.: The Role of Knowledge on Genetic Counseling and Testing in Black Cancer Survivors at Increased Risk of Carrying a BRCA1/2 Mutation. J Genet Couns 26 (1): 113-121, 2017. [PUBMED Abstract]
  45. Cragun D, Weidner A, Lewis C, et al.: Racial disparities in BRCA testing and cancer risk management across a population-based sample of young breast cancer survivors. Cancer 123 (13): 2497-2505, 2017. [PUBMED Abstract]
  46. Cragun D, Weidner A, Kechik J, et al.: Genetic Testing Across Young Hispanic and Non-Hispanic White Breast Cancer Survivors: Facilitators, Barriers, and Awareness of the Genetic Information Nondiscrimination Act. Genet Test Mol Biomarkers 23 (2): 75-83, 2019. [PUBMED Abstract]
  47. Lerman C, Hughes C, Lemon SJ, et al.: What you don't know can hurt you: adverse psychologic effects in members of BRCA1-linked and BRCA2-linked families who decline genetic testing. J Clin Oncol 16 (5): 1650-4, 1998. [PUBMED Abstract]
  48. Lodder L, Frets PG, Trijsburg RW, et al.: Attitudes and distress levels in women at risk to carry a BRCA1/BRCA2 gene mutation who decline genetic testing. Am J Med Genet 119A (3): 266-72, 2003. [PUBMED Abstract]
  49. Foster C, Evans DG, Eeles R, et al.: Non-uptake of predictive genetic testing for BRCA1/2 among relatives of known carriers: attributes, cancer worry, and barriers to testing in a multicenter clinical cohort. Genet Test 8 (1): 23-9, 2004. [PUBMED Abstract]
  50. McInerney-Leo A, Biesecker BB, Hadley DW, et al.: BRCA1/2 testing in hereditary breast and ovarian cancer families II: impact on relationships. Am J Med Genet A 133 (2): 165-9, 2005. [PUBMED Abstract]
  51. Patenaude AF: Cancer susceptibility testing: risks, benefits, and personal beliefs. In: Clarke A, ed.: The Genetic Testing of Children. BIOS Scientific, 1998, pp 145-156.
  52. Richards M: The genetic testing of children: adult attitude's and children's understanding. In: Clarke A, ed.: The Genetic Testing of Children. BIOS Scientific, 1998, pp 169-179.
  53. Wertz DC, Fanos JH, Reilly PR: Genetic testing for children and adolescents. Who decides? JAMA 272 (11): 875-81, 1994. [PUBMED Abstract]
  54. Borry P, Stultiëns L, Nys H, et al.: Attitudes towards predictive genetic testing in minors for familial breast cancer: a systematic review. Crit Rev Oncol Hematol 64 (3): 173-81, 2007. [PUBMED Abstract]
  55. Hamann HA, Croyle RT, Venne VL, et al.: Attitudes toward the genetic testing of children among adults in a Utah-based kindred tested for a BRCA1 mutation. Am J Med Genet 92 (1): 25-32, 2000. [PUBMED Abstract]
  56. Bradbury AR, Patrick-Miller L, Pawlowski K, et al.: Should genetic testing for BRCA1/2 be permitted for minors? Opinions of BRCA mutation carriers and their adult offspring. Am J Med Genet C Semin Med Genet 148C (1): 70-7, 2008. [PUBMED Abstract]
  57. Points to consider: ethical, legal, and psychosocial implications of genetic testing in children and adolescents. American Society of Human Genetics Board of Directors, American College of Medical Genetics Board of Directors. Am J Hum Genet 57 (5): 1233-41, 1995. [PUBMED Abstract]
  58. Michie S, Marteau TM: Predictive genetic testing in children: the need for psychological research. In: Clarke A, ed.: The Genetic Testing of Children. BIOS Scientific, 1998, pp 169-182.
  59. MacDonald DJ, Lessick M: Hereditary cancers in children and ethical and psychosocial implications. J Pediatr Nurs 15 (4): 217-25, 2000. [PUBMED Abstract]
  60. Tercyak KP, Peshkin BN, Streisand R, et al.: Psychological issues among children of hereditary breast cancer gene (BRCA1/2) testing participants. Psychooncology 10 (4): 336-46, 2001 Jul-Aug. [PUBMED Abstract]
  61. Winer E, Winer N, Bluman L, et al.: Attitudes and risk perceptions of women with breast cancer considering testing for BRCA1/2. [Abstract] Proceedings of the American Society of Clinical Oncology 16: A1937, 537a, 1997.
  62. Braithwaite D, Emery J, Walter F, et al.: Psychological impact of genetic counseling for familial cancer: a systematic review and meta-analysis. J Natl Cancer Inst 96 (2): 122-33, 2004. [PUBMED Abstract]
  63. Mikkelsen EM, Sunde L, Johansen C, et al.: Psychosocial conditions of women awaiting genetic counseling: a population-based study. J Genet Couns 17 (3): 242-51, 2008. [PUBMED Abstract]
  64. Dorval M, Bouchard K, Maunsell E, et al.: Health behaviors and psychological distress in women initiating BRCA1/2 genetic testing: comparison with control population. J Genet Couns 17 (4): 314-26, 2008. [PUBMED Abstract]
  65. Wang C, Gonzalez R, Janz N, et al.: The role of cognitive appraisal and worry in BRCA1/2 testing decisions among a clinic population. Psychol Health 22 (6): 719-36, 2007.
  66. Hallowell N, Statham H, Murton F: Women's understanding of their risk of developing breast/ovarian cancer before and after genetic counseling. J Genet Couns 7 (4): 345-64, 1998.
  67. MacDonald DJ, Choi J, Ferrell B, et al.: Concerns of women presenting to a comprehensive cancer centre for genetic cancer risk assessment. J Med Genet 39 (7): 526-30, 2002. [PUBMED Abstract]
  68. Matloff ET, Moyer A, Shannon KM, et al.: Healthy women with a family history of breast cancer: impact of a tailored genetic counseling intervention on risk perception, knowledge, and menopausal therapy decision making. J Womens Health (Larchmt) 15 (7): 843-56, 2006. [PUBMED Abstract]
  69. Meiser B, Price MA, Butow PN, et al.: Misperceptions of ovarian cancer risk in women at increased risk for hereditary ovarian cancer. Fam Cancer 13 (2): 153-62, 2014. [PUBMED Abstract]
  70. Bluman LG, Rimer BK, Berry DA, et al.: Attitudes, knowledge, and risk perceptions of women with breast and/or ovarian cancer considering testing for BRCA1 and BRCA2. J Clin Oncol 17 (3): 1040-6, 1999. [PUBMED Abstract]
  71. Iglehart JD, Miron A, Rimer BK, et al.: Overestimation of hereditary breast cancer risk. Ann Surg 228 (3): 375-84, 1998. [PUBMED Abstract]
  72. Bluman LG, Rimer BK, Regan Sterba K, et al.: Attitudes, knowledge, risk perceptions and decision-making among women with breast and/or ovarian cancer considering testing for BRCA1 and BRCA2 and their spouses. Psychooncology 12 (5): 410-27, 2003 Jul-Aug. [PUBMED Abstract]
  73. McCaul KD, O'Donnell SM: Naive beliefs about breast cancer risk. Womens Health 4 (1): 93-101, 1998 Spring. [PUBMED Abstract]
  74. Huiart L, Eisinger F, Stoppa-Lyonnet D, et al.: Effects of genetic consultation on perception of a family risk of breast/ovarian cancer and determinants of inaccurate perception after the consultation. J Clin Epidemiol 55 (7): 665-75, 2002. [PUBMED Abstract]
  75. Davis S, Stewart S, Bloom J: Increasing the accuracy of perceived breast cancer risk: results from a randomized trial with Cancer Information Service callers. Prev Med 39 (1): 64-73, 2004. [PUBMED Abstract]
  76. Katapodi MC, Dodd MJ, Lee KA, et al.: Underestimation of breast cancer risk: influence on screening behavior. Oncol Nurs Forum 36 (3): 306-14, 2009. [PUBMED Abstract]
  77. Lindberg NM, Wellisch D: Anxiety and compliance among women at high risk for breast cancer. Ann Behav Med 23 (4): 298-303, 2001 Fall. [PUBMED Abstract]
  78. Ritvo P, Irvine J, Robinson G, et al.: Psychological adjustment to familial-genetic risk assessment for ovarian cancer: predictors of nonadherence to surveillance recommendations. Gynecol Oncol 84 (1): 72-80, 2002. [PUBMED Abstract]
  79. Meiser B, Halliday JL: What is the impact of genetic counselling in women at increased risk of developing hereditary breast cancer? A meta-analytic review. Soc Sci Med 54 (10): 1463-70, 2002. [PUBMED Abstract]
  80. Green J, Richards M, Murton F, et al.: Family communication and genetic counseling: the case of hereditary breast and ovarian cancer. J Genet Couns 6 (1): 45-60, 1997.
  81. Quillin JM, Ramakrishnan V, Borzelleca J, et al.: Paternal relatives and family history of breast cancer. Am J Prev Med 31 (3): 265-8, 2006. [PUBMED Abstract]
  82. O'Neill SM, Rubinstein WS, Wang C, et al.: Familial risk for common diseases in primary care: the Family Healthware Impact Trial. Am J Prev Med 36 (6): 506-14, 2009. [PUBMED Abstract]
  83. Rubinstein WS, O'neill SM, Rothrock N, et al.: Components of family history associated with women's disease perceptions for cancer: a report from the Family Healthware™ Impact Trial. Genet Med 13 (1): 52-62, 2011. [PUBMED Abstract]
  84. Theis B, Boyd N, Lockwood G, et al.: Accuracy of family cancer history in breast cancer patients. Eur J Cancer Prev 3 (4): 321-7, 1994. [PUBMED Abstract]
  85. Breuer B, Kash KM, Rosenthal G, et al.: Reporting bilaterality status in first-degree relatives with breast cancer: a validity study. Genet Epidemiol 10 (4): 245-56, 1993. [PUBMED Abstract]
  86. Parent ME, Ghadirian P, Lacroix A, et al.: The reliability of recollections of family history: implications for the medical provider. J Cancer Educ 12 (2): 114-20, 1997 Summer. [PUBMED Abstract]
  87. Kelly KM, Shedlosky-Shoemaker R, Porter K, et al.: Cancer family history reporting: impact of method and psychosocial factors. J Genet Couns 16 (3): 373-82, 2007. [PUBMED Abstract]
  88. Kerber RA, Slattery ML: Comparison of self-reported and database-linked family history of cancer data in a case-control study. Am J Epidemiol 146 (3): 244-8, 1997. [PUBMED Abstract]
  89. Kerr B, Foulkes WD, Cade D, et al.: False family history of breast cancer in the family cancer clinic. Eur J Surg Oncol 24 (4): 275-9, 1998. [PUBMED Abstract]
  90. Schwartz MD, Peshkin BN, Hughes C, et al.: Impact of BRCA1/BRCA2 mutation testing on psychologic distress in a clinic-based sample. J Clin Oncol 20 (2): 514-20, 2002. [PUBMED Abstract]
  91. Mancini J, Noguès C, Adenis C, et al.: Impact of an information booklet on satisfaction and decision-making about BRCA genetic testing. Eur J Cancer 42 (7): 871-81, 2006. [PUBMED Abstract]
  92. Green MJ, Biesecker BB, McInerney AM, et al.: An interactive computer program can effectively educate patients about genetic testing for breast cancer susceptibility. Am J Med Genet 103 (1): 16-23, 2001. [PUBMED Abstract]
  93. Green MJ, Peterson SK, Baker MW, et al.: Effect of a computer-based decision aid on knowledge, perceptions, and intentions about genetic testing for breast cancer susceptibility: a randomized controlled trial. JAMA 292 (4): 442-52, 2004. [PUBMED Abstract]
  94. Green MJ, McInerney AM, Biesecker BB, et al.: Education about genetic testing for breast cancer susceptibility: patient preferences for a computer program or genetic counselor. Am J Med Genet 103 (1): 24-31, 2001. [PUBMED Abstract]
  95. Dabney MK, Huelsman K: Counseling by computer: breast cancer risk and genetic testing. Developed by the University of Wisconsin-Madison Department of Medicine and the Program in Medical Ethics. Genet Test 4 (1): 43-4, 2000. [PUBMED Abstract]
  96. Green MJ, Peterson SK, Baker MW, et al.: Use of an educational computer program before genetic counseling for breast cancer susceptibility: effects on duration and content of counseling sessions. Genet Med 7 (4): 221-9, 2005. [PUBMED Abstract]
  97. Wang C, Gonzalez R, Milliron KJ, et al.: Genetic counseling for BRCA1/2: a randomized controlled trial of two strategies to facilitate the education and counseling process. Am J Med Genet A 134 (1): 66-73, 2005. [PUBMED Abstract]
  98. Joseph G, Beattie MS, Lee R, et al.: Pre-counseling education for low literacy women at risk of Hereditary Breast and Ovarian Cancer (HBOC): patient experiences using the Cancer Risk Education Intervention Tool (CREdIT). J Genet Couns 19 (5): 447-62, 2010. [PUBMED Abstract]
  99. Albada A, van Dulmen S, Lindhout D, et al.: A pre-visit tailored website enhances counselees' realistic expectations and knowledge and fulfils information needs for breast cancer genetic counselling. Fam Cancer 11 (1): 85-95, 2012. [PUBMED Abstract]
  100. Baty BJ, Kinney AY, Ellis SM: Developing culturally sensitive cancer genetics communication aids for African Americans. Am J Med Genet 118A (2): 146-55, 2003. [PUBMED Abstract]
  101. Permuth-Wey J, Vadaparampil S, Rumphs A, et al.: Development of a culturally tailored genetic counseling booklet about hereditary breast and ovarian cancer for Black women. Am J Med Genet A 152A (4): 836-45, 2010. [PUBMED Abstract]
  102. Pal T, Stowe C, Cole A, et al.: Evaluation of phone-based genetic counselling in African American women using culturally tailored visual aids. Clin Genet 78 (2): 124-31, 2010. [PUBMED Abstract]
  103. Calzone KA: Predisposition testing for breast and ovarian cancer susceptibility. Semin Oncol Nurs 13 (2): 82-90, 1997. [PUBMED Abstract]
  104. Smith KR, West JA, Croyle RT, et al.: Familial context of genetic testing for cancer susceptibility: moderating effect of siblings' test results on psychological distress one to two weeks after BRCA1 mutation testing. Cancer Epidemiol Biomarkers Prev 8 (4 Pt 2): 385-92, 1999. [PUBMED Abstract]
  105. Wylie JE, Smith KR, Botkin JR: Effects of spouses on distress experienced by BRCA1 mutation carriers over time. Am J Med Genet 119C (1): 35-44, 2003. [PUBMED Abstract]
  106. Kash KM, Holland JC, Halper MS, et al.: Psychological distress and surveillance behaviors of women with a family history of breast cancer. J Natl Cancer Inst 84 (1): 24-30, 1992. [PUBMED Abstract]
  107. Lerman C, Schwartz M: Adherence and psychological adjustment among women at high risk for breast cancer. Breast Cancer Res Treat 28 (2): 145-55, 1993. [PUBMED Abstract]
  108. Kelly PT: Understanding Breast Cancer Risk. Temple University Press, 1991.
  109. Baty BJ, Venne VL, McDonald J, et al.: BRCA1 testing: genetic counseling protocol development and counseling issues. J Genet Couns 6 (2): 223-44, 1997.
  110. Richards MP, Hallowell N, Green JM, et al.: Counseling families with hereditary breast and ovarian cancer: a psychosocial perspective. J Genet Couns 4 (3): 219-33, 1995.
  111. Hoskins KF, Stopfer JE, Calzone KA, et al.: Assessment and counseling for women with a family history of breast cancer. A guide for clinicians. JAMA 273 (7): 577-85, 1995. [PUBMED Abstract]
  112. Schneider KA: Genetic counseling for BRCA1/BRCA2 testing. Genet Test 1 (2): 91-8, 1997. [PUBMED Abstract]
  113. McKinnon WC, Baty BJ, Bennett RL, et al.: Predisposition genetic testing for late-onset disorders in adults. A position paper of the National Society of Genetic Counselors. JAMA 278 (15): 1217-20, 1997. [PUBMED Abstract]
  114. Cummings S, Olopade O: Predisposition testing for inherited breast cancer. Oncology (Huntingt) 12 (8): 1227-41; discussion 1241-2, 1998. [PUBMED Abstract]
  115. Lipkus IM, Klein WM, Rimer BK: Communicating breast cancer risks to women using different formats. Cancer Epidemiol Biomarkers Prev 10 (8): 895-8, 2001. [PUBMED Abstract]
  116. Butow PN, Lobb EA: Analyzing the process and content of genetic counseling in familial breast cancer consultations. J Genet Couns 13 (5): 403-24, 2004. [PUBMED Abstract]
  117. Lerman C, Audrain J, Croyle RT: DNA-testing for heritable breast cancer risks: lessons from traditional genetic counseling. Ann Behav Med 16(4): 327-333, 1994.
  118. Pieterse AH, van Dulmen AM, Beemer FA, et al.: Cancer genetic counseling: communication and counselees' post-visit satisfaction, cognitions, anxiety, and needs fulfillment. J Genet Couns 16 (1): 85-96, 2007. [PUBMED Abstract]
  119. Hamilton JG, Lobel M, Moyer A: Emotional distress following genetic testing for hereditary breast and ovarian cancer: a meta-analytic review. Health Psychol 28 (4): 510-8, 2009. [PUBMED Abstract]
  120. Beran TM, Stanton AL, Kwan L, et al.: The trajectory of psychological impact in BRCA1/2 genetic testing: does time heal? Ann Behav Med 36 (2): 107-16, 2008. [PUBMED Abstract]
  121. Bosch N, Junyent N, Gadea N, et al.: What factors may influence psychological well being at three months and one year post BRCA genetic result disclosure? Breast 21 (6): 755-60, 2012. [PUBMED Abstract]
  122. O'Neill SC, Rini C, Goldsmith RE, et al.: Distress among women receiving uninformative BRCA1/2 results: 12-month outcomes. Psychooncology 18 (10): 1088-96, 2009. [PUBMED Abstract]
  123. Oberguggenberger A, Sztankay M, Morscher RJ, et al.: Psychosocial outcomes and counselee satisfaction following genetic counseling for hereditary breast and ovarian cancer: A patient-reported outcome study. J Psychosom Res 89: 39-45, 2016. [PUBMED Abstract]
  124. Foster C, Watson M, Eeles R, et al.: Predictive genetic testing for BRCA1/2 in a UK clinical cohort: three-year follow-up. Br J Cancer 96 (5): 718-24, 2007. [PUBMED Abstract]
  125. Halbert CH, Stopfer JE, McDonald J, et al.: Long-term reactions to genetic testing for BRCA1 and BRCA2 mutations: does time heal women's concerns? J Clin Oncol 29 (32): 4302-6, 2011. [PUBMED Abstract]
  126. Graves KD, Vegella P, Poggi EA, et al.: Long-term psychosocial outcomes of BRCA1/BRCA2 testing: differences across affected status and risk-reducing surgery choice. Cancer Epidemiol Biomarkers Prev 21 (3): 445-55, 2012. [PUBMED Abstract]
  127. Cella D, Hughes C, Peterman A, et al.: A brief assessment of concerns associated with genetic testing for cancer: the Multidimensional Impact of Cancer Risk Assessment (MICRA) questionnaire. Health Psychol 21 (6): 564-72, 2002. [PUBMED Abstract]
  128. Shkedi-Rafid S, Gabai-Kapara E, Grinshpun-Cohen J, et al.: BRCA genetic testing of individuals from families with low prevalence of cancer: experiences of carriers and implications for population screening. Genet Med 14 (7): 688-94, 2012. [PUBMED Abstract]
  129. Metcalfe KA, Poll A, Llacuachaqui M, et al.: Patient satisfaction and cancer-related distress among unselected Jewish women undergoing genetic testing for BRCA1 and BRCA2. Clin Genet 78 (5): 411-7, 2010. [PUBMED Abstract]
  130. Metcalfe KA, Mian N, Enmore M, et al.: Long-term follow-up of Jewish women with a BRCA1 and BRCA2 mutation who underwent population genetic screening. Breast Cancer Res Treat 133 (2): 735-40, 2012. [PUBMED Abstract]
  131. Armstrong J, Toscano M, Kotchko N, et al.: Utilization and Outcomes of BRCA Genetic Testing and Counseling in a National Commercially Insured Population: The ABOUT Study. JAMA Oncol 1 (9): 1251-60, 2015. [PUBMED Abstract]
  132. Dohany L, Gustafson S, Ducaine W, et al.: Psychological distress with direct-to-consumer genetic testing: a case report of an unexpected BRCA positive test result. J Genet Couns 21 (3): 399-401, 2012. [PUBMED Abstract]
  133. Mahon SM: Impact of direct-to-consumer genetic testing. J Oncol Pract 8 (4): 260, 2012. [PUBMED Abstract]
  134. Jeffers L, Morrison PJ, McCaughan E, et al.: Maximising survival: the main concern of women with hereditary breast and ovarian cancer who undergo genetic testing for BRCA1/2. Eur J Oncol Nurs 18 (4): 411-8, 2014. [PUBMED Abstract]
  135. Francke U, Dijamco C, Kiefer AK, et al.: Dealing with the unexpected: consumer responses to direct-access BRCA mutation testing. PeerJ 1: e8, 2013. [PUBMED Abstract]
  136. Meiser B, Quinn VF, Gleeson M, et al.: When knowledge of a heritable gene mutation comes out of the blue: treatment-focused genetic testing in women newly diagnosed with breast cancer. Eur J Hum Genet 24 (11): 1517-1523, 2016. [PUBMED Abstract]
  137. Ardern-Jones A, Kenen R, Eeles R: Too much, too soon? Patients and health professionals' views concerning the impact of genetic testing at the time of breast cancer diagnosis in women under the age of 40. Eur J Cancer Care (Engl) 14 (3): 272-81, 2005. [PUBMED Abstract]
  138. Wevers MR, Hahn DE, Verhoef S, et al.: Breast cancer genetic counseling after diagnosis but before treatment: a pilot study on treatment consequences and psychological impact. Patient Educ Couns 89 (1): 89-95, 2012. [PUBMED Abstract]
  139. Zilliacus E, Meiser B, Gleeson M, et al.: Are we being overly cautious? A qualitative inquiry into the experiences and perceptions of treatment-focused germline BRCA genetic testing amongst women recently diagnosed with breast cancer. Support Care Cancer 20 (11): 2949-58, 2012. [PUBMED Abstract]
  140. Tercyak KP, Peshkin BN, Brogan BM, et al.: Quality of life after contralateral prophylactic mastectomy in newly diagnosed high-risk breast cancer patients who underwent BRCA1/2 gene testing. J Clin Oncol 25 (3): 285-91, 2007. [PUBMED Abstract]
  141. Wevers MR, Ausems MG, Verhoef S, et al.: Does rapid genetic counseling and testing in newly diagnosed breast cancer patients cause additional psychosocial distress? results from a randomized clinical trial. Genet Med 18 (2): 137-44, 2016. [PUBMED Abstract]
  142. Wevers MR, Aaronson NK, Bleiker EMA, et al.: Rapid genetic counseling and testing in newly diagnosed breast cancer: Patients' and health professionals' attitudes, experiences, and evaluation of effects on treatment decision making. J Surg Oncol 116 (8): 1029-1039, 2017. [PUBMED Abstract]
  143. Lerman C, Peshkin BN, Hughes C, et al.: Family disclosure in genetic testing for cancer susceptibility: determinants and consequences. Journal of Health Care Law and Policy 1 (2): 353-73, 1998.
  144. Elrick A, Ashida S, Ivanovich J, et al.: Psychosocial and Clinical Factors Associated with Family Communication of Cancer Genetic Test Results among Women Diagnosed with Breast Cancer at a Young Age. J Genet Couns 26 (1): 173-181, 2017. [PUBMED Abstract]
  145. Finlay E, Stopfer JE, Burlingame E, et al.: Factors determining dissemination of results and uptake of genetic testing in families with known BRCA1/2 mutations. Genet Test 12 (1): 81-91, 2008. [PUBMED Abstract]
  146. Hughes C, Lerman C, Schwartz M, et al.: All in the family: evaluation of the process and content of sisters' communication about BRCA1 and BRCA2 genetic test results. Am J Med Genet 107 (2): 143-50, 2002. [PUBMED Abstract]
  147. Baars JE, Ausems MG, van Riel E, et al.: Communication Between Breast Cancer Patients Who Received Inconclusive Genetic Test Results and Their Daughters and Sisters Years After Testing. J Genet Couns 25 (3): 461-71, 2016. [PUBMED Abstract]
  148. Wagner Costalas J, Itzen M, Malick J, et al.: Communication of BRCA1 and BRCA2 results to at-risk relatives: a cancer risk assessment program's experience. Am J Med Genet 119C (1): 11-8, 2003. [PUBMED Abstract]
  149. Patenaude AF, Dorval M, DiGianni LS, et al.: Sharing BRCA1/2 test results with first-degree relatives: factors predicting who women tell. J Clin Oncol 24 (4): 700-6, 2006. [PUBMED Abstract]
  150. MacDonald DJ, Sarna L, van Servellen G, et al.: Selection of family members for communication of cancer risk and barriers to this communication before and after genetic cancer risk assessment. Genet Med 9 (5): 275-82, 2007. [PUBMED Abstract]
  151. Kenen R, Arden-Jones A, Eeles R: Healthy women from suspected hereditary breast and ovarian cancer families: the significant others in their lives. Eur J Cancer Care (Engl) 13 (2): 169-79, 2004. [PUBMED Abstract]
  152. Claes E, Evers-Kiebooms G, Boogaerts A, et al.: Communication with close and distant relatives in the context of genetic testing for hereditary breast and ovarian cancer in cancer patients. Am J Med Genet 116A (1): 11-9, 2003. [PUBMED Abstract]
  153. Foster C, Eeles R, Ardern-Jones A, et al.: Juggling roles and expectations: dilemmas faced by women talking to relatives about cancer and genetic testing. Psychol Health 19 (4): 439-55, 2004.
  154. Kenen R, Arden-Jones A, Eeles R: We are talking, but are they listening? Communication patterns in families with a history of breast/ovarian cancer (HBOC). Psychooncology 13 (5): 335-45, 2004. [PUBMED Abstract]
  155. Segal J, Esplen MJ, Toner B, et al.: An investigation of the disclosure process and support needs of BRCA1 and BRCA2 carriers. Am J Med Genet A 125 (3): 267-72, 2004. [PUBMED Abstract]
  156. Bradbury AR, Dignam JJ, Ibe CN, et al.: How often do BRCA mutation carriers tell their young children of the family's risk for cancer? A study of parental disclosure of BRCA mutations to minors and young adults. J Clin Oncol 25 (24): 3705-11, 2007. [PUBMED Abstract]
  157. Bradbury AR, Patrick-Miller L, Pawlowski K, et al.: Learning of your parent's BRCA mutation during adolescence or early adulthood: a study of offspring experiences. Psychooncology 18 (2): 200-8, 2009. [PUBMED Abstract]
  158. Manne S, Audrain J, Schwartz M, et al.: Associations between relationship support and psychological reactions of participants and partners to BRCA1 and BRCA2 testing in a clinic-based sample. Ann Behav Med 28 (3): 211-25, 2004. [PUBMED Abstract]
  159. Daly MB, Montgomery S, Bingler R, et al.: Communicating genetic test results within the family: Is it lost in translation? A survey of relatives in the randomized six-step study. Fam Cancer 15 (4): 697-706, 2016. [PUBMED Abstract]
  160. McAllister MF, Evans DG, Ormiston W, et al.: Men in breast cancer families: a preliminary qualitative study of awareness and experience. J Med Genet 35 (9): 739-44, 1998. [PUBMED Abstract]
  161. Liede A, Metcalfe K, Hanna D, et al.: Evaluation of the needs of male carriers of mutations in BRCA1 or BRCA2 who have undergone genetic counseling. Am J Hum Genet 67 (6): 1494-504, 2000. [PUBMED Abstract]
  162. Metcalfe KA, Liede A, Trinkaus M, et al.: Evaluation of the needs of spouses of female carriers of mutations in BRCA1 and BRCA2. Clin Genet 62 (6): 464-9, 2002. [PUBMED Abstract]
  163. Mireskandari S, Sherman KA, Meiser B, et al.: Psychological adjustment among partners of women at high risk of developing breast/ovarian cancer. Genet Med 9 (5): 311-20, 2007. [PUBMED Abstract]
  164. Sherman KA, Kasparian NA, Mireskandari S: Psychological adjustment among male partners in response to women's breast/ovarian cancer risk: a theoretical review of the literature. Psychooncology 19 (1): 1-11, 2010. [PUBMED Abstract]
  165. Daly MB: The impact of social roles on the experience of men in BRCA1/2 families: implications for counseling. J Genet Couns 18 (1): 42-8, 2009. [PUBMED Abstract]
  166. DudokdeWit AC, Tibben A, Frets PG, et al.: Males at-risk for the BRCA1 gene, the psychological impact. Psychooncology 5(3): 251-257, 1996.
  167. Lodder L, Frets PG, Trijsburg RW, et al.: Men at risk of being a mutation carrier for hereditary breast/ovarian cancer: an exploration of attitudes and psychological functioning during genetic testing. Eur J Hum Genet 9 (7): 492-500, 2001. [PUBMED Abstract]
  168. Lerman C, Hughes C, Croyle RT, et al.: Prophylactic surgery decisions and surveillance practices one year following BRCA1/2 testing. Prev Med 31 (1): 75-80, 2000. [PUBMED Abstract]
  169. Hughes C, Lynch H, Durham C, et al.: Communication of BRCA1/2 Test Results in Hereditary Breast Cancer Families. Cancer Research in Therapy and Control Vol. 8, 1999, pp. 51-59.
  170. Tercyak KP, Hughes C, Main D, et al.: Parental communication of BRCA1/2 genetic test results to children. Patient Educ Couns 42 (3): 213-24, 2001. [PUBMED Abstract]
  171. Tercyak KP, Peshkin BN, DeMarco TA, et al.: Parent-child factors and their effect on communicating BRCA1/2 test results to children. Patient Educ Couns 47 (2): 145-53, 2002. [PUBMED Abstract]
  172. McGivern B, Everett J, Yager GG, et al.: Family communication about positive BRCA1 and BRCA2 genetic test results. Genet Med 6 (6): 503-9, 2004 Nov-Dec. [PUBMED Abstract]
  173. Bradbury AR, Patrick-Miller L, Schwartz L, et al.: Psychosocial Adjustment in School-age Girls With a Family History of Breast Cancer. Pediatrics 136 (5): 927-37, 2015. [PUBMED Abstract]
  174. Bradbury AR, Patrick-Miller L, Schwartz LA, et al.: Psychosocial Adjustment and Perceived Risk Among Adolescent Girls From Families With BRCA1/2 or Breast Cancer History. J Clin Oncol 34 (28): 3409-16, 2016. [PUBMED Abstract]
  175. Tercyak KP, Peshkin BN, Demarco TA, et al.: Information needs of mothers regarding communicating BRCA1/2 cancer genetic test results to their children. Genet Test 11 (3): 249-55, 2007. [PUBMED Abstract]
  176. Wertz DC: International perspectives. In: Clarke A, ed.: The Genetic Testing of Children. BIOS Scientific, 1998, pp 271-287.
  177. Benkendorf JL, Reutenauer JE, Hughes CA, et al.: Patients' attitudes about autonomy and confidentiality in genetic testing for breast-ovarian cancer susceptibility. Am J Med Genet 73 (3): 296-303, 1997. [PUBMED Abstract]
  178. Staton AD, Kurian AW, Cobb K, et al.: Cancer risk reduction and reproductive concerns in female BRCA1/2 mutation carriers. Fam Cancer 7 (2): 179-86, 2008. [PUBMED Abstract]
  179. Friedman LC, Kramer RM: Reproductive issues for women with BRCA mutations. J Natl Cancer Inst Monogr (34): 83-6, 2005. [PUBMED Abstract]
  180. Smith KR, Ellington L, Chan AY, et al.: Fertility intentions following testing for a BRCA1 gene mutation. Cancer Epidemiol Biomarkers Prev 13 (5): 733-40, 2004. [PUBMED Abstract]
  181. Quinn G, Vadaparampil S, Wilson C, et al.: Attitudes of high-risk women toward preimplantation genetic diagnosis. Fertil Steril 91 (6): 2361-8, 2009. [PUBMED Abstract]
  182. Cunniff C; American Academy of Pediatrics Committee on Genetics: Prenatal screening and diagnosis for pediatricians. Pediatrics 114 (3): 889-94, 2004. [PUBMED Abstract]
  183. Rappaport VJ: Prenatal diagnosis and genetic screening--integration into prenatal care. Obstet Gynecol Clin North Am 35 (3): 435-58, ix, 2008. [PUBMED Abstract]
  184. Baruch S, Kaufman D, Hudson KL: Genetic testing of embryos: practices and perspectives of US in vitro fertilization clinics. Fertil Steril 89 (5): 1053-8, 2008. [PUBMED Abstract]
  185. Ogilvie CM, Braude PR, Scriven PN: Preimplantation genetic diagnosis--an overview. J Histochem Cytochem 53 (3): 255-60, 2005. [PUBMED Abstract]
  186. Vadaparampil ST, Quinn GP, Knapp C, et al.: Factors associated with preimplantation genetic diagnosis acceptance among women concerned about hereditary breast and ovarian cancer. Genet Med 11 (10): 757-65, 2009. [PUBMED Abstract]
  187. Quinn GP, Vadaparampil ST, King LM, et al.: Conflict between values and technology: perceptions of preimplantation genetic diagnosis among women at increased risk for hereditary breast and ovarian cancer. Fam Cancer 8 (4): 441-9, 2009. [PUBMED Abstract]
  188. Chan JL, Johnson LNC, Sammel MD, et al.: Reproductive Decision-Making in Women with BRCA1/2 Mutations. J Genet Couns 26 (3): 594-603, 2017. [PUBMED Abstract]
  189. Menon U, Harper J, Sharma A, et al.: Views of BRCA gene mutation carriers on preimplantation genetic diagnosis as a reproductive option for hereditary breast and ovarian cancer. Hum Reprod 22 (6): 1573-7, 2007. [PUBMED Abstract]
  190. Fortuny D, Balmaña J, Graña B, et al.: Opinion about reproductive decision making among individuals undergoing BRCA1/2 genetic testing in a multicentre Spanish cohort. Hum Reprod 24 (4): 1000-6, 2009. [PUBMED Abstract]
  191. Julian-Reynier C, Fabre R, Coupier I, et al.: BRCA1/2 carriers: their childbearing plans and theoretical intentions about having preimplantation genetic diagnosis and prenatal diagnosis. Genet Med 14 (5): 527-34, 2012. [PUBMED Abstract]
  192. Ormondroyd E, Donnelly L, Moynihan C, et al.: Attitudes to reproductive genetic testing in women who had a positive BRCA test before having children: a qualitative analysis. Eur J Hum Genet 20 (1): 4-10, 2012. [PUBMED Abstract]
  193. Quinn GP, Vadaparampil ST, Miree CA, et al.: High risk men's perceptions of pre-implantation genetic diagnosis for hereditary breast and ovarian cancer. Hum Reprod 25 (10): 2543-50, 2010. [PUBMED Abstract]
  194. Struewing JP, Abeliovich D, Peretz T, et al.: The carrier frequency of the BRCA1 185delAG mutation is approximately 1 percent in Ashkenazi Jewish individuals. Nat Genet 11 (2): 198-200, 1995. [PUBMED Abstract]
  195. Rothenberg KH: Breast cancer, the genetic "quick fix," and the Jewish community. Ethical, legal, and social challenges. Health Matrix Clevel 7 (1): 97-124, 1997 Winter. [PUBMED Abstract]
  196. Foster MW, Bernsten D, Carter TH: A model agreement for genetic research in socially identifiable populations. Am J Hum Genet 63 (3): 696-702, 1998. [PUBMED Abstract]
  197. Burhansstipanov L, Bemis LT, Dignan MB: Native American cancer education: genetic and cultural issues. J Cancer Educ 16 (3): 142-5, 2001 Autumn. [PUBMED Abstract]
  198. Hughes C, Fasaye GA, LaSalle VH, et al.: Sociocultural influences on participation in genetic risk assessment and testing among African American women. Patient Educ Couns 51 (2): 107-14, 2003. [PUBMED Abstract]
  199. Julian-Reynier CM, Bouchard LJ, Evans DG, et al.: Women's attitudes toward preventive strategies for hereditary breast or ovarian carcinoma differ from one country to another: differences among English, French, and Canadian women. Cancer 92 (4): 959-68, 2001. [PUBMED Abstract]
  200. Phillips KA, Warner E, Meschino WS, et al.: Perceptions of Ashkenazi Jewish breast cancer patients on genetic testing for mutations in BRCA1 and BRCA2. Clin Genet 57 (5): 376-83, 2000. [PUBMED Abstract]
  201. Vadaparampil ST, Quinn GP, Small BJ, et al.: A pilot study of hereditary breast and ovarian knowledge among a multiethnic group of Hispanic women with a personal or family history of cancer. Genet Test Mol Biomarkers 14 (1): 99-106, 2010. [PUBMED Abstract]
  202. Halbert CH, Kessler L, Troxel AB, et al.: Effect of genetic counseling and testing for BRCA1 and BRCA2 mutations in African American women: a randomized trial. Public Health Genomics 13 (7-8): 440-8, 2010. [PUBMED Abstract]
  203. Freedman TG: Genetic susceptibility testing: ethical and social quandaries. Health Soc Work 23 (3): 214-22, 1998. [PUBMED Abstract]
  204. Hubbard R, Lewontin RC: Pitfalls of genetic testing. N Engl J Med 334 (18): 1192-4, 1996. [PUBMED Abstract]
  205. Parens E: Glad and terrified: on the ethics of BRACA1 and 2 testing. Cancer Invest 14 (4): 405-11, 1996. [PUBMED Abstract]
  206. Winter PR, Wiesner GL, Finnegan J, et al.: Notification of a family history of breast cancer: issues of privacy and confidentiality. Am J Med Genet 66 (1): 1-6, 1996. [PUBMED Abstract]
  207. Geller G, Doksum T, Bernhardt BA, et al.: Participation in breast cancer susceptibility testing protocols: influence of recruitment source, altruism, and family involvement on women's decisions. Cancer Epidemiol Biomarkers Prev 8 (4 Pt 2): 377-83, 1999. [PUBMED Abstract]
  208. Rimer BK, Schildkraut JM, Lerman C, et al.: Participation in a women's breast cancer risk counseling trial. Who participates? Who declines? High Risk Breast Cancer Consortium. Cancer 77 (11): 2348-55, 1996. [PUBMED Abstract]
  209. Robson ME, Storm CD, Weitzel J, et al.: American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol 28 (5): 893-901, 2010. [PUBMED Abstract]
  210. Burke W, Daly M, Garber J, et al.: Recommendations for follow-up care of individuals with an inherited predisposition to cancer. II. BRCA1 and BRCA2. Cancer Genetics Studies Consortium. JAMA 277 (12): 997-1003, 1997. [PUBMED Abstract]
  211. Durfy SJ, Buchanan TE, Burke W: Testing for inherited susceptibility to breast cancer: a survey of informed consent forms for BRCA1 and BRCA2 mutation testing. Am J Med Genet 75 (1): 82-7, 1998. [PUBMED Abstract]
  212. Statement of the American Society of Clinical Oncology: genetic testing for cancer susceptibility, Adopted on February 20, 1996. J Clin Oncol 14 (5): 1730-6; discussion 1737-40, 1996. [PUBMED Abstract]
  213. Hallowell N, Foster C, Eeles R, et al.: Balancing autonomy and responsibility: the ethics of generating and disclosing genetic information. J Med Ethics 29 (2): 74-9; discussion 80-3, 2003. [PUBMED Abstract]
  214. Metcalfe KA, Poll A, O'Connor A, et al.: Development and testing of a decision aid for breast cancer prevention for women with a BRCA1 or BRCA2 mutation. Clin Genet 72 (3): 208-17, 2007. [PUBMED Abstract]
  215. Tiller K, Meiser B, Gaff C, et al.: A randomized controlled trial of a decision aid for women at increased risk of ovarian cancer. Med Decis Making 26 (4): 360-72, 2006 Jul-Aug. [PUBMED Abstract]
  216. 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 22 (16): 3293-301, 2004. [PUBMED Abstract]
  217. Culver JO, MacDonald DJ, Thornton AA, et al.: Development and evaluation of a decision aid for BRCA carriers with breast cancer. J Genet Couns 20 (3): 294-307, 2011. [PUBMED Abstract]
  218. Metcalfe KA, Dennis CL, Poll A, et al.: Effect of decision aid for breast cancer prevention on decisional conflict in women with a BRCA1 or BRCA2 mutation: a multisite, randomized, controlled trial. Genet Med 19 (3): 330-336, 2017. [PUBMED Abstract]
  219. Schwartz MD, Lerman C, Brogan B, et al.: Impact of BRCA1/BRCA2 counseling and testing on newly diagnosed breast cancer patients. J Clin Oncol 22 (10): 1823-9, 2004. [PUBMED Abstract]
  220. Botkin JR, Smith KR, Croyle RT, et al.: Genetic testing for a BRCA1 mutation: prophylactic surgery and screening behavior in women 2 years post testing. Am J Med Genet A 118 (3): 201-9, 2003. [PUBMED Abstract]
  221. Beattie MS, Crawford B, Lin F, et al.: Uptake, time course, and predictors of risk-reducing surgeries in BRCA carriers. Genet Test Mol Biomarkers 13 (1): 51-6, 2009. [PUBMED Abstract]
  222. O'Neill SC, Valdimarsdottir HB, Demarco TA, et al.: BRCA1/2 test results impact risk management attitudes, intentions, and uptake. Breast Cancer Res Treat 124 (3): 755-64, 2010. [PUBMED Abstract]
  223. Schwartz MD, Isaacs C, Graves KD, et al.: Long-term outcomes of BRCA1/BRCA2 testing: risk reduction and surveillance. Cancer 118 (2): 510-7, 2012. [PUBMED Abstract]
  224. Garcia C, Wendt J, Lyon L, et al.: Risk management options elected by women after testing positive for a BRCA mutation. Gynecol Oncol 132 (2): 428-33, 2014. [PUBMED Abstract]
  225. Singh K, Lester J, Karlan B, et al.: Impact of family history on choosing risk-reducing surgery among BRCA mutation carriers. Am J Obstet Gynecol 208 (4): 329.e1-6, 2013. [PUBMED Abstract]
  226. Phillips KA, Jenkins MA, Lindeman GJ, et al.: Risk-reducing surgery, screening and chemoprevention practices of BRCA1 and BRCA2 mutation carriers: a prospective cohort study. Clin Genet 70 (3): 198-206, 2006. [PUBMED Abstract]
  227. Metcalfe KA, Birenbaum-Carmeli D, Lubinski J, et al.: International variation in rates of uptake of preventive options in BRCA1 and BRCA2 mutation carriers. Int J Cancer 122 (9): 2017-22, 2008. [PUBMED Abstract]
  228. Julian-Reynier C, Mancini J, Mouret-Fourme E, et al.: Cancer risk management strategies and perceptions of unaffected women 5 years after predictive genetic testing for BRCA1/2 mutations. Eur J Hum Genet 19 (5): 500-6, 2011. [PUBMED Abstract]
  229. 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 20 (5): 1260-8, 2002. [PUBMED Abstract]
  230. Mannis GN, Fehniger JE, Creasman JS, et al.: Risk-reducing salpingo-oophorectomy and ovarian cancer screening in 1077 women after BRCA testing. JAMA Intern Med 173 (2): 96-103, 2013. [PUBMED Abstract]
  231. Friebel TM, Domchek SM, Neuhausen SL, et al.: Bilateral prophylactic oophorectomy and bilateral prophylactic mastectomy in a prospective cohort of unaffected BRCA1 and BRCA2 mutation carriers. Clin Breast Cancer 7 (11): 875-82, 2007. [PUBMED Abstract]
  232. Madalinska JB, van Beurden M, Bleiker EM, et al.: Predictors of prophylactic bilateral salpingo-oophorectomy compared with gynecologic screening use in BRCA1/2 mutation carriers. J Clin Oncol 25 (3): 301-7, 2007. [PUBMED Abstract]
  233. Rhiem K, Foth D, Wappenschmidt B, et al.: Risk-reducing salpingo-oophorectomy in BRCA1 and BRCA2 mutation carriers. Arch Gynecol Obstet 283 (3): 623-7, 2011. [PUBMED Abstract]
  234. Sidon L, Ingham S, Clancy T, et al.: Uptake of risk-reducing salpingo-oophorectomy in women carrying a BRCA1 or BRCA2 mutation: evidence for lower uptake in women affected by breast cancer and older women. Br J Cancer 106 (4): 775-9, 2012. [PUBMED Abstract]
  235. Lerman C, Narod S, Schulman K, et al.: BRCA1 testing in families with hereditary breast-ovarian cancer. A prospective study of patient decision making and outcomes. JAMA 275 (24): 1885-92, 1996. [PUBMED Abstract]
  236. van Dijk S, van Roosmalen MS, Otten W, et al.: Decision making regarding prophylactic mastectomy: stability of preferences and the impact of anticipated feelings of regret. J Clin Oncol 26 (14): 2358-63, 2008. [PUBMED Abstract]
  237. Claes E, Evers-Kiebooms G, Decruyenaere M, et al.: Surveillance behavior and prophylactic surgery after predictive testing for hereditary breast/ovarian cancer. Behav Med 31 (3): 93-105, 2005. [PUBMED Abstract]
  238. Ray JA, Loescher LJ, Brewer M: Risk-reduction surgery decisions in high-risk women seen for genetic counseling. J Genet Couns 14 (6): 473-84, 2005. [PUBMED Abstract]
  239. Hallowell N: 'You don't want to lose your ovaries because you think 'I might become a man". Women's perceptions of prophylactic surgery as a cancer risk management option. Psychooncology 7 (3): 263-75, 1998 May-Jun. [PUBMED Abstract]
  240. Schneider KA, Stopfer JE, Peters JA, et al.: Complexities in cancer risk counseling: presentation of three cases. J Genet Couns 6 (2): 147-67, 1997.
  241. Tarkan L: My Mother's Breast: Daughters Face Their Mothers' Cancer. Taylor Publishing, 1999.
  242. Stefanek ME, Helzlsouer KJ, Wilcox PM, et al.: Predictors of and satisfaction with bilateral prophylactic mastectomy. Prev Med 24 (4): 412-9, 1995. [PUBMED Abstract]
  243. Cortesi L, Razzaboni E, Toss A, et al.: A rapid genetic counselling and testing in newly diagnosed breast cancer is associated with high rate of risk-reducing mastectomy in BRCA1/2-positive Italian women. Ann Oncol 25 (1): 57-63, 2014. [PUBMED Abstract]
  244. Rosenberg SM, Ruddy KJ, Tamimi RM, et al.: BRCA1 and BRCA2 Mutation Testing in Young Women With Breast Cancer. JAMA Oncol 2 (6): 730-6, 2016. [PUBMED Abstract]
  245. Graves KD, Peshkin BN, Halbert CH, et al.: Predictors and outcomes of contralateral prophylactic mastectomy among breast cancer survivors. Breast Cancer Res Treat 104 (3): 321-9, 2007. [PUBMED Abstract]
  246. Howard-McNatt M, Schroll RW, Hurt GJ, et al.: Contralateral prophylactic mastectomy in breast cancer patients who test negative for BRCA mutations. Am J Surg 202 (3): 298-302, 2011. [PUBMED Abstract]
  247. Bresser PJ, Seynaeve C, Van Gool AR, et al.: Satisfaction with prophylactic mastectomy and breast reconstruction in genetically predisposed women. Plast Reconstr Surg 117 (6): 1675-82; discussion 1683-4, 2006. [PUBMED Abstract]
  248. Brandberg Y, Sandelin K, Erikson S, et al.: Psychological reactions, quality of life, and body image after bilateral prophylactic mastectomy in women at high risk for breast cancer: a prospective 1-year follow-up study. J Clin Oncol 26 (24): 3943-9, 2008. [PUBMED Abstract]
  249. Lobb E, Meiser B: Genetic counselling and prophylactic surgery in women from families with hereditary breast or ovarian cancer. Lancet 363 (9424): 1841-2, 2004. [PUBMED Abstract]
  250. Lobb EA, Butow PN, Meiser B, et al.: Tailoring communication in consultations with women from high risk breast cancer families. Br J Cancer 87 (5): 502-8, 2002. [PUBMED Abstract]
  251. Lobb EA, Butow PN, Barratt A, et al.: Communication and information-giving in high-risk breast cancer consultations: influence on patient outcomes. Br J Cancer 90 (2): 321-7, 2004. [PUBMED Abstract]
  252. Schwartz MD, Kaufman E, Peshkin BN, et al.: Bilateral prophylactic oophorectomy and ovarian cancer screening following BRCA1/BRCA2 mutation testing. J Clin Oncol 21 (21): 4034-41, 2003. [PUBMED Abstract]
  253. Kauff ND, Satagopan JM, Robson ME, et al.: Risk-reducing salpingo-oophorectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med 346 (21): 1609-15, 2002. [PUBMED Abstract]
  254. Schmeler KM, Sun CC, Bodurka DC, et al.: Prophylactic bilateral salpingo-oophorectomy compared with surveillance in women with BRCA mutations. Obstet Gynecol 108 (3 Pt 1): 515-20, 2006. [PUBMED Abstract]
  255. MacDonald DJ, Sarna L, Uman GC, et al.: Cancer screening and risk-reducing behaviors of women seeking genetic cancer risk assessment for breast and ovarian cancers. Oncol Nurs Forum 33 (2): E27-35, 2006. [PUBMED Abstract]
  256. Litton JK, Westin SN, Ready K, et al.: Perception of screening and risk reduction surgeries in patients tested for a BRCA deleterious mutation. Cancer 115 (8): 1598-604, 2009. [PUBMED Abstract]
  257. Tyndel S, Austoker J, Henderson BJ, et al.: What is the psychological impact of mammographic screening on younger women with a family history of breast cancer? Findings from a prospective cohort study by the PIMMS Management Group. J Clin Oncol 25 (25): 3823-30, 2007. [PUBMED Abstract]
  258. Rees G, Young MA, Gaff C, et al.: A qualitative study of health professionals' views regarding provision of information about health-protective behaviors during genetic consultation for breast cancer. J Genet Couns 15 (2): 95-104, 2006. [PUBMED Abstract]
  259. Lodder LN, Frets PG, Trijsburg RW, et al.: One year follow-up of women opting for presymptomatic testing for BRCA1 and BRCA2: emotional impact of the test outcome and decisions on risk management (surveillance or prophylactic surgery). Breast Cancer Res Treat 73 (2): 97-112, 2002. [PUBMED Abstract]
  260. van Oostrom I, Meijers-Heijboer H, Lodder LN, et al.: Long-term psychological impact of carrying a BRCA1/2 mutation and prophylactic surgery: a 5-year follow-up study. J Clin Oncol 21 (20): 3867-74, 2003. [PUBMED Abstract]
  261. Bresser PJ, Seynaeve C, Van Gool AR, et al.: The course of distress in women at increased risk of breast and ovarian cancer due to an (identified) genetic susceptibility who opt for prophylactic mastectomy and/or salpingo-oophorectomy. Eur J Cancer 43 (1): 95-103, 2007. [PUBMED Abstract]
  262. Frost MH, Schaid DJ, Sellers TA, et al.: Long-term satisfaction and psychological and social function following bilateral prophylactic mastectomy. JAMA 284 (3): 319-24, 2000. [PUBMED Abstract]
  263. Metcalfe KA, Esplen MJ, Goel V, et al.: Psychosocial functioning in women who have undergone bilateral prophylactic mastectomy. Psychooncology 13 (1): 14-25, 2004. [PUBMED Abstract]
  264. Schlich-Bakker KJ, Ausems MG, Schipper M, et al.: BRCA1/2 mutation testing in breast cancer patients: a prospective study of the long-term psychological impact of approach during adjuvant radiotherapy. Breast Cancer Res Treat 109 (3): 507-14, 2008. [PUBMED Abstract]
  265. Frost MH, Slezak JM, Tran NV, et al.: Satisfaction after contralateral prophylactic mastectomy: the significance of mastectomy type, reconstructive complications, and body appearance. J Clin Oncol 23 (31): 7849-56, 2005. [PUBMED Abstract]
  266. Schwartz MD: Contralateral prophylactic mastectomy: efficacy, satisfaction, and regret. J Clin Oncol 23 (31): 7777-9, 2005. [PUBMED Abstract]
  267. Metcalfe KA, Cil TD, Semple JL, et al.: Long-Term Psychosocial Functioning in Women with Bilateral Prophylactic Mastectomy: Does Preservation of the Nipple-Areolar Complex Make a Difference? Ann Surg Oncol 22 (10): 3324-30, 2015. [PUBMED Abstract]
  268. Yao K, Liederbach E, Tang R, et al.: Nipple-sparing mastectomy in BRCA1/2 mutation carriers: an interim analysis and review of the literature. Ann Surg Oncol 22 (2): 370-6, 2015. [PUBMED Abstract]
  269. Geiger AM, Nekhlyudov L, Herrinton LJ, et al.: Quality of life after bilateral prophylactic mastectomy. Ann Surg Oncol 14 (2): 686-94, 2007. [PUBMED Abstract]
  270. Isern AE, Tengrup I, Loman N, et al.: Aesthetic outcome, patient satisfaction, and health-related quality of life in women at high risk undergoing prophylactic mastectomy and immediate breast reconstruction. J Plast Reconstr Aesthet Surg 61 (10): 1177-87, 2008. [PUBMED Abstract]
  271. Kenen RH, Shapiro PJ, Hantsoo L, et al.: Women with BRCA1 or BRCA2 mutations renegotiating a post-prophylactic mastectomy identity: self-image and self-disclosure. J Genet Couns 16 (6): 789-98, 2007. [PUBMED Abstract]
  272. Altschuler A, Nekhlyudov L, Rolnick SJ, et al.: Positive, negative, and disparate--women's differing long-term psychosocial experiences of bilateral or contralateral prophylactic mastectomy. Breast J 14 (1): 25-32, 2008 Jan-Feb. [PUBMED Abstract]
  273. Patenaude AF, Orozco S, Li X, et al.: Support needs and acceptability of psychological and peer consultation: attitudes of 108 women who had undergone or were considering prophylactic mastectomy. Psychooncology 17 (8): 831-43, 2008. [PUBMED Abstract]
  274. Elit L, Esplen MJ, Butler K, et al.: Quality of life and psychosexual adjustment after prophylactic oophorectomy for a family history of ovarian cancer. Fam Cancer 1 (3-4): 149-56, 2001. [PUBMED Abstract]
  275. Robson M, Hensley M, Barakat R, et al.: Quality of life in women at risk for ovarian cancer who have undergone risk-reducing oophorectomy. Gynecol Oncol 89 (2): 281-7, 2003. [PUBMED Abstract]
  276. Finch A, Metcalfe KA, Chiang JK, et al.: The impact of prophylactic salpingo-oophorectomy on menopausal symptoms and sexual function in women who carry a BRCA mutation. Gynecol Oncol 121 (1): 163-8, 2011. [PUBMED Abstract]
  277. Finch A, Metcalfe K, Lui J, et al.: Breast and ovarian cancer risk perception after prophylactic salpingo-oophorectomy due to an inherited mutation in the BRCA1 or BRCA2 gene. Clin Genet 75 (3): 220-4, 2009. [PUBMED Abstract]
  278. Madalinska JB, Hollenstein J, Bleiker E, et al.: Quality-of-life effects of prophylactic salpingo-oophorectomy versus gynecologic screening among women at increased risk of hereditary ovarian cancer. J Clin Oncol 23 (28): 6890-8, 2005. [PUBMED Abstract]
  279. Westin SN, Sun CC, Lu KH, et al.: Satisfaction with ovarian carcinoma risk-reduction strategies among women at high risk for breast and ovarian carcinoma. Cancer 117 (12): 2659-67, 2011. [PUBMED Abstract]
  280. Campfield Bonadies D, Moyer A, Matloff ET: What I wish I'd known before surgery: BRCA carriers' perspectives after bilateral salipingo-oophorectomy. Fam Cancer 10 (1): 79-85, 2011. [PUBMED Abstract]
  281. Isaacs C, Peshkin BN, Schwartz M, et al.: Breast and ovarian cancer screening practices in healthy women with a strong family history of breast or ovarian cancer. Breast Cancer Res Treat 71 (2): 103-12, 2002. [PUBMED Abstract]
  282. Peshkin BN, Schwartz MD, Isaacs C, et al.: Utilization of breast cancer screening in a clinically based sample of women after BRCA1/2 testing. Cancer Epidemiol Biomarkers Prev 11 (10 Pt 1): 1115-8, 2002. [PUBMED Abstract]
  283. Tinley ST, Houfek J, Watson P, et al.: Screening adherence in BRCA1/2 families is associated with primary physicians' behavior. Am J Med Genet A 125 (1): 5-11, 2004. [PUBMED Abstract]
  284. Lerman C, Seay J, Balshem A, et al.: Interest in genetic testing among first-degree relatives of breast cancer patients. Am J Med Genet 57 (3): 385-92, 1995. [PUBMED Abstract]
  285. Struewing JP, Lerman C, Kase RG, et al.: Anticipated uptake and impact of genetic testing in hereditary breast and ovarian cancer families. Cancer Epidemiol Biomarkers Prev 4 (2): 169-73, 1995. [PUBMED Abstract]
  286. Jacobsen PB, Valdimarsdottier HB, Brown KL, et al.: Decision-making about genetic testing among women at familial risk for breast cancer. Psychosom Med 59 (5): 459-66, 1997 Sep-Oct. [PUBMED Abstract]
  287. Watson M, Kash KM, Homewood J, et al.: Does genetic counseling have any impact on management of breast cancer risk? Genet Test 9 (2): 167-74, 2005. [PUBMED Abstract]

Changes to This Summary (04/07/2022)

The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.

Introduction

Updated National Comprehensive Cancer Network (NCCN) (Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic) as reference 153.

High-Penetrance Breast and/or Gynecologic Cancer Susceptibility Genes

Updated NCCN (Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic) as reference 99.

Clinical Management of Carriers of BRCA Pathogenic Variants

Updated Table 13, Available Clinical Practice Guidelines for Hereditary Breast and Ovarian Cancer, with NCCN guidelines (cited as NCCN [Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic] as reference 33).

Psychosocial Issues in BRCA1/BRCA2

This section was previously named Psychosocial Issues in Inherited Breast and Ovarian Cancer Syndromes.

This summary is written and maintained by the PDQ Cancer Genetics Editorial Board, which is editorially independent of NCI. The summary reflects an independent review of the literature and does not represent a policy statement of NCI or NIH. More information about summary policies and the role of the PDQ Editorial Boards in maintaining the PDQ summaries can be found on the About This PDQ Summary and PDQ® - NCI's Comprehensive Cancer Database pages.

About This PDQ Summary

Purpose of This Summary

This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the genetics of breast and gynecologic cancers. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.

Reviewers and Updates

This summary is reviewed regularly and updated as necessary by the PDQ Cancer Genetics Editorial Board, which is editorially independent of the National Cancer Institute (NCI). The summary reflects an independent review of the literature and does not represent a policy statement of NCI or the National Institutes of Health (NIH).

Board members review recently published articles each month to determine whether an article should:

  • be discussed at a meeting,
  • be cited with text, or
  • replace or update an existing article that is already cited.

Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.

The lead reviewers for Genetics of Breast and Gynecologic Cancers are:

  • Doreen Agnese, MD (The Ohio State University)
  • Kathleen A. Calzone, PhD, RN, AGN-BC, FAAN (National Cancer Institute)
  • Ilana Cass, MD (Dartmouth-Hitchcock Medical Center)
  • Lee-may Chen, MD (UCSF Helen Diller Family Comprehensive Cancer Center)
  • Mary B. Daly, MD, PhD (Fox Chase Cancer Center)
  • Tuya Pal, MD, FACMG, FCCMG (Vanderbilt-Ingram Cancer Center)
  • Beth N. Peshkin, MS, CGC (Lombardi Comprehensive Cancer Center at Georgetown University)
  • Susan K. Peterson, PhD, MPH (University of Texas, M.D. Anderson Cancer Center)
  • John M. Quillin, PhD, MPH, MS (Virginia Commonwealth University)
  • Mary Beth Terry, PhD (Columbia University Mailman School of Public Health)
  • Susan T. Vadaparampil, PhD, MPH (H. Lee Moffitt Cancer Center & Research Institute)
  • Catharine Wang, PhD, MSc (Boston University School of Public Health)

Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's Email Us. Do not contact the individual Board Members with questions or comments about the summaries. Board members will not respond to individual inquiries.

Levels of Evidence

Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Cancer Genetics Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.

Permission to Use This Summary

PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as “NCI’s PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary].”

The preferred citation for this PDQ summary is:

PDQ® Cancer Genetics Editorial Board. PDQ Genetics of Breast and Gynecologic Cancers. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at: https://www.cancer.gov/types/breast/hp/breast-ovarian-genetics-pdq. Accessed <MM/DD/YYYY>. [PMID: 26389210]

Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in Visuals Online, a collection of over 2,000 scientific images.

Disclaimer

The information in these summaries should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the Managing Cancer Care page.

Contact Us

More information about contacting us or receiving help with the Cancer.gov website can be found on our Contact Us for Help page. Questions can also be submitted to Cancer.gov through the website’s Email Us.

Updated:

This content is provided by the National Cancer Institute (www.cancer.gov)
Syndicated Content Details:
Source URL: https://www.cancer.gov/node/2569/syndication
Source Agency: National Cancer Institute (NCI)
Captured Date: 2013-09-14 09:01:04.0