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.    
            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             LS, 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 LS.          
            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.          
                        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 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.          
      [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. 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) database. When OMIM appears after a gene name or the      name of a condition, click on OMIM for a link to more      information.]    
      Among women, 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 2016, an      estimated 249,260 new cases will be diagnosed, and 40,890      deaths from breast cancer will occur.[1] The incidence of breast cancer,      particularly for estrogen receptorpositive 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 Womens Health Initiative (WHI).[2] An estimated 22,280 new cases of      ovarian cancer are expected in 2016, with an estimated 14,240      deaths. Ovarian cancer is the fifth most deadly cancer in      women.[1] An estimated 60,050      new cases of endometrial cancer are expected in 2016, with an      estimated 10,470 deaths.[1]      (Refer to the PDQ summaries on Breast Cancer      Treatment; 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.    
      Refer to the PDQ summary on Breast Cancer      Prevention for information about risk factors for breast      cancer in the general population.    
      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.[3-6]      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.[7] 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], 2.02.2).[7] 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.[4,5,7-9] 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, 1.81.9)      and was 2.7 (95% CI, 2.62.9) 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, 3.14.8). 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.[9] (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.)    
      Cumulative risk of breast cancer increases with age, with      most breast cancers occurring after age 50 years.[10] In women with a       genetic susceptibility, breast cancer, and to a lesser      degree, ovarian cancer, tends to occur at an earlier age than      in sporadic cases.    
      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      years), with a more significant effect seen for carriers of      BRCA1 pathogenic variants.[11-13] Likewise, breast feeding can      reduce breast cancer risk in carriers of BRCA1 (but      not BRCA2) pathogenic variants.[14] 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.[15] 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.[16]    
      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 (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.[17]    
      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.[18] However, use of      OCs formulated before 1975 was associated with an increased      risk of breast cancer (summary relative risk [SRR], 1.47; 95%      CI, 1.062.04).[18] (Refer to      the Reproductive      factors section in the Clinical      Management of Carriers of BRCA Pathogenic Variants      section of this summary for more information.)    
      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, 1.211.49) for women who had used HRT for 5 or more      years after menopause.[19]      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.[20,21] 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, 1.021.5, 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 Womens Study in the United Kingdom.[22] 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, 0.591.01). 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.[23]    
      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.[24-28,19] 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.[28] 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.[21] Short-term use of hormones for      treatment of menopausal symptoms appears to confer little or      no breast cancer risk.[19,29] The effect of HRT on breast      cancer risk among carriers of BRCA1 or      BRCA2 pathogenic variants has been studied only 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.[30] (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.)    
      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,[31-34] and in association with germline      ATM and TP53 variants.[35,36]    
      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      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.[37] 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.[38-43] 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.[42] 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.[43] (Refer to the Mammography      section in the Clinical      Management of Carriers of BRCA Pathogenic Variants      section of this summary for more information about      radiation.)    
      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.[44,45] Prior studies of carriers of      BRCA1/BRCA2 pathogenic variants have found no      increased risk associated with alcohol consumption.[46,47]    
      Weight gain and being overweight are commonly recognized risk      factors for breast cancer. In general, overweight women are      most commonly observed to be at increased risk of      postmenopausal breast cancer and at reduced risk of      premenopausal breast cancer. Sedentary lifestyle may also be      a risk factor.[48] These      factors have not been systematically evaluated in women with      a positive family history of breast cancer or in carriers of      cancer-predisposing pathogenic variants, but one study      suggested a reduced risk of cancer associated with exercise      among carriers of BRCA1 and BRCA2      pathogenic variants.[49]    
      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).[50] There may also be an association      between BBD and family history of breast cancer.[51]    
      An increased risk of breast cancer has also been demonstrated      for women who have increased density of breast tissue as      assessed by mammogram,[50,52,53] and breast density is likely to      have a genetic component in its etiology.[54-56]    
      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 [57] 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.[58]    
      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.    
      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.[59]    
      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.[60]    
      Nulliparity is consistently associated with an increased risk      of ovarian cancer, including among carriers of      BRCA/BRCA2 pathogenic variants, yet a meta-analysis      could only identify risk-reduction in women with four or more      live births.[13] Risk may      also be increased among women who have used fertility drugs,      especially those who remain nulligravid.[61,62] Several studies have reported a      risk reduction in ovarian cancer after OC pill use in      carriers of BRCA1/BRCA2 pathogenic      variants;[63-65] 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.[65,66] 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.[67-70]    
      Bilateral tubal ligation and hysterectomy are associated with      reduced ovarian cancer risk,[61,71,72] including in carriers of      BRCA1/BRCA2 pathogenic variants.[73] Ovarian cancer risk is reduced      more than 90% in women with documented BRCA1 or      BRCA2 pathogenic variants who chose risk-reducing      salpingo-oophorectomy. In this same population, risk-reducing      oophorectomy also resulted in a nearly 50% reduction in the      risk of subsequent breast cancer.[74,75] (Refer to the Risk-reducing      salpingo-oophorectomy section of this summary for more      information about these studies.)    
      Use of OCs for 4 or more years is associated with an      approximately 50% reduction in ovarian cancer risk in the      general population.[61,76] A majority of, but not all,      studies also support OCs being protective among carriers of      BRCA1/BRCA2 pathogenic variants.[66,77-80] 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, 0.330.75) associated with OC      use.[18] (Refer to the      Oral      contraceptives section in the Chemoprevention      section of this summary for more information.)    
      Refer to the PDQ summary on Endometrial      Cancer Prevention for information about risk factors for      endometrial cancer in the general population.    
      Although the hyperestrogenic state is the most common      predisposing factor for endometrial cancer, family history      also plays a significant role in a womans risk for disease.      Approximately 3% to 5% of uterine cancer cases are      attributable to a hereditary cause,[81] with the main hereditary      endometrial cancer syndrome being Lynch syndrome (LS), an      autosomal dominant genetic condition with a population      prevalence of 1 in 300 to 1 in 1,000 individuals.[82,83] (Refer to the LS      section in the PDQ summary on Genetics of      Colorectal Cancer for more information.)    
      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.[84] Women with      LS tend to develop endometrial cancer at an earlier age, with      the median age at diagnosis of 48 years.[85]    
      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.[86,87]    
      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 (PCOS), 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.[88,89] 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.[90] In contrast, oral      contraceptives, 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.[91-94]    
      Autosomal dominant inheritance of breast and gynecologic      cancers is characterized by transmission of cancer       predisposition from generation to generation, through      either the mothers or the fathers side of the family, with      the following characteristics:    
      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.[95] 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      LS,      basal cell nevus (Gorlin) syndrome (OMIM), and      multiple endocrine neoplasia type 1 (OMIM).[95] LS is mainly associated with      colorectal cancer and endometrial cancer, although several      studies have demonstrated that patients with LS 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).[96-102]    
            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:    
      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      LS. (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.)    
      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.    
      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.    
      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 and a younger age at onset in some      individuals. 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.    
      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 receptornegative, HER2/neu      receptornegative (i.e., triple-negative breast cancers), 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      LS-associated pathogenic variants from noncarriers. The      hallmark feature of endometrial cancers occurring in LS is      mismatch repair (MMR) defects, 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.    
      The accuracy and completeness of family histories must be      taken into account 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%.[103,104] 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 to predict an individuals lifetime risk of developing      breast and/or gynecologic cancer are available.[105-108] In addition, models exist to      predict an individuals likelihood of having a pathogenic      variant in BRCA1, BRCA2, or one of the MMR      genes associated with LS. (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 patients 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.[106,109,110]    
      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.[110] 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]) [111], and the Colditz and Rosner      model [112]) require only      limited information about family history (e.g., number of      first-degree relatives 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 [113]), others assuming two loci      (e.g., the International Breast Cancer Intervention Study      [IBIS] model [114] and the      BRCAPRO model [115]), 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 [116-118]). The models also differ in      whether they include information about nongenetic risk      factors. Three models (Gail/BCRAT, Pfeiffer,[108] 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).[119] The existing models generally      are more accurate in prospective studies that have assessed      how well they predict future cancers.[110,120-122]    
      In the United States, BRCAPRO, the Claus model,[113,123] and the Gail/BCRAT [111] 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.    
      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.[124-129] Risk can be overestimated in      the following populations:    
      The Gail/BCRAT model is valid for women aged 35 years and      older. The model was primarily developed for white      women.[128] 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.[130] Additionally, extensions of the      Gail model have incorporated high-risk       single nucleotide polymorphisms and pathogenic variants;      however, no software exists to calculate risk in these      extended models.[131,132]      Other risk assessment models incorporating breast density      have been developed but are not ready for clinical      use.[133,134]    
      Generally, the Gail/BCRAT model should not be the sole model      used for families with one or more of the following      characteristics:    
      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.[114] 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.[117] 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,[116-118] which is more in line with what      is known about the underlying genetics of breast cancer.      However, the discrimination and calibration for these models      differ significantly when compared in independent      samples;[120] the IBIS and      BOADICEA models are more comparable when estimating risk over      a shorter fixed time horizon (e.g., 10 years),[120] 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 womans 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.    
      Two risk predictions models have been developed for ovarian      cancer.[107,108] The Rosner model [107] included age at menopause, age      at menarche, oral contraception use, and tubal ligation; the      concordance statistic was 0.60 (0.570.62). The Pfeiffer      model [108] 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 (0.560.62). Although both      models were well calibrated, their modest discriminatory      power limited their screening potential.    
      The Pfeiffer model has been used to predict endometrial      cancer risk in the general population.[108] For endometrial cancer, the      relative risk model included BMI, menopausal hormone therapy      use, menopausal status, age at menopause, smoking status, and      oral contraceptive pill use. The discriminatory power of the      model was 0.68 (0.660.70); 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, PREMM1,2,6, and MMRpro      are three quantitative predictive models used to identify      individuals who may potentially have LS.[135-137] MMRpredict incorporates only      colorectal cancer patients but does include MSI and      immunohistochemistry (IHC) tumor testing results.      PREMM1,2,6 accounts for other LS-associated tumors      but does not include tumor testing results. 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.[138] However,      because these models were developed and validated in      colorectal cancer patients, the discriminative abilities of      these models to identify LS are lower among individuals with      endometrial cancer than among those with colon      cancer.[139] 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 LS 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.    
      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.    
      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 (HR), and risk      ratio.    
      Absolute risk measures take into account 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), and 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]    
      Figure 5. These five pedigrees depict probands with varying      degrees of family history. Table 2 accompanies this figure.    
      With the increasing use of multigene panel tests (see            below), 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.    
      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 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 due to 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.[1] In a      population of ovarian cancer patients who test negative for      BRCA1 and BRCA2       pathogenic variants, multigene panel testing can reveal      actionable pathogenic variants.[2,3] 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 cancerpredisposing genes      was 4.6%.[4] A caveat is the      possible finding of a       variant of uncertain significance, where the clinical      significance remains unknown. 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.)    
      Epidemiologic studies have clearly established the role of            family history as an important risk factor for both      breast and ovarian cancer. 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 some of these      cancer 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.    
      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 (OMIM) 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.    
      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 (OMIM) 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 only and in up to 90% of      families with both breast and ovarian cancer.[16]    
      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, persons 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, and      as part of, 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]    
      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 risk of cancer 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, or large deletions or       insertions are missed by most of the techniques,      including direct DNA sequencing, but testing for these is      commercially available. Such rearrangements are believed to      be responsible for 12% to 18% of BRCA1 inactivating      variants but are less frequently seen in BRCA2 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]    
      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 but will have a            variant of uncertain (or unknown) significance (VUS). VUS      may cause substantial challenges in 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 patients 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 over a 3-year      period.[32] Among subjects      who had no 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 with no clear pathogenic variants had sequence      alterations that were once considered VUS but were      reclassified as a       polymorphism, or occasionally as a pathogenic variant.    
      The frequency of VUS varies by ethnicity within the U.S.      population. African Americans 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 track VUS in the family to      determine if there is       cosegregation of the VUS with the cancer in the 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 in itself deleterious, although there      are rare exceptions. As an adjunct to the clinical      information, models to interpret VUS have been developed,      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.    
      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 apparently 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 Americans and Hispanics.[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. For example, it is estimated that up to      15% of BRCA1 and BRCA2 pathogenic variants      that occur among Ashkenazim are nonfounder pathogenic      variants.[32]    
      Among the general population, the likelihood of having any      BRCA variant is as follows:    
      Among AJ individuals, the likelihood of having any      BRCA variant is as follows:    
      Two large U.S. population-based studies of breast cancer      patients younger than age 65 years examined the prevalence of      BRCA1 [55,70] and BRCA2 [55] pathogenic variants in various      ethnic groups. The prevalence of BRCA1 pathogenic      variants in breast cancer patients by ethnic group was 3.5%      in Hispanics, 1.3% to 1.4% in African Americans, 0.5% in      Asian Americans, 2.2% to 2.9% in non-AJ whites, and 8.3% to      10.2% in AJ individuals.[55,70] The prevalence of BRCA2      pathogenic variants by ethnic group was 2.6% in African      Americans and 2.1% in whites.[55]    
      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);[71] 21 of the 189 (11%) BRCA      pathogenic variants identified were large rearrangements, of      which 13 (62%) were the BRCA1 exon 912 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 912      deletion.[72] In another      population-based cohort of 492 Hispanic women with breast      cancer, the BRCA1 exon 912 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%.[69] 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).[73] 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.[6,69,73,74]    
      Population screening has identified carriers in a number of      AJ populations who would not have met criteria for      family-based testing.[62,75-77] This could potentially expand the      number of individuals who could benefit from preventive      strategies. 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.    
      Several studies have assessed the frequency of BRCA1      or BRCA2 pathogenic variants in women with breast or      ovarian cancer.[55,56,70,78-86] Personal characteristics      associated with an increased likelihood of a BRCA1      and/or BRCA2 pathogenic variant include the      following:    
      Family history characteristics associated with an increased      likelihood of carrying a BRCA1 and/or BRCA2      pathogenic variant include the following:    
      Several professional organizations and expert panels,      including the American Society of Clinical Oncology,[91] the National Comprehensive Cancer      Network (NCCN),[92] the      American Society of Human Genetics,[93] the American College of Medical      Genetics and Genomics,[94]      the National Society of Genetic Counselors,[94] the U.S. Preventive Services Task      Force,[95] and the Society of      Gynecologic Oncologists,[96]      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.    
      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,[32,78,79,81,84,97,98] genetic models using Bayesian      analysis (BRCAPRO and Breast and Ovarian Analysis of Disease      Incidence and Carrier Estimation Algorithm      [BOADICEA]),[84,99] and empiric      observations,[52,55,58,100-102] including the       Myriad prevalence tables.    
      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 (see Table      3). The discrimination and accuracy (factors used to      evaluate the performance of prediction models) of these      models are much higher for these models' ability to report on      carrier status than for their ability to predict fixed or      remaining lifetime risk.    
Read the original here:
Genetics of Breast and Gynecologic Cancers (PDQ)Health ...