Abstract
Background
It has become increasingly common to consider BRCA mutation status when determining optimal cancer risk management and treatment options in order to improve patient outcomes. Knowledge about the risk for hereditary cancer at or as close as possible to the time of diagnosis allows patients access to the most risk reduction options available.
Methods
This paper illustrates the role of genetic risk assessment for hereditary breast cancer, using hereditary breast and ovarian cancer (HBOC) syndrome as a model due to germline mutations in the BRCA1 and BRCA2. Specifically, the value of genetic counseling and testing for HBOC across the cancer prevention and control continuum is outlined as it pertains to breast cancer.
Results
In recognition of the importance of risk assessment for hereditary breast cancer, leading health professional organizations have developed specific guidelines and recommendations to providers for identification of women at increased risk for carrying a BRCA mutation.
Conclusions
Institutional efforts specific to genetic counseling and testing have resulted in the implementation of a model driven by physician recommendation as a referral system for high-risk breast cancer patients. Establishing an infrastructure to support research, education, and outreach initiatives focused on BRCA genetic counseling and testing will provide information that can improve the delivery of cancer genetics services.
Overview of Genetic Counseling and Testing Using BRCA as a Model
The majority of inherited breast cancers are attributed to hereditary breast and ovarian cancer (HBOC) syndrome due to mutations in the BRCA1 and BRCA2 (BRCA) genes. Although there are several other genes that also lead to hereditary breast cancer predisposition (eg, PTEN, P53, STK11), they are much rarer. Furthermore, for the purposes of this review, we will not specifically consider individuals at high risk by virtue of a striking family history, in whom a germline BRCA mutation is not identified (recognizing that in the clinical setting, it may be appropriate to offer these individuals many of the risk management options recommended for BRCA mutation carriers). Consequently, we use HBOC as a model for outlining the importance of genetic counseling and testing services across the cancer prevention and control continuum (Figure).
Figure.

Management of inherited breast cancer across the cancer prevention and control continuum.
The BRCA genes were discovered approximately 15 years ago,1,2 representing a tremendous opportunity to identify individuals at greatly elevated risk for HBOC.1–3 Mutations in these genes account for approximately 5% of all breast cancers4 and 10% to 15% of all ovarian cancers.2,5,6 Lifetime cancer risks to age 70 in BRCA mutation carriers are approximately 60% to 70% for breast cancer7–11 and 40% or lower for ovarian cancer.7,9 Moreover, the risk of a second primary breast cancer in BRCA mutation carriers, particularly those diagnosed with breast cancer at a younger age, is much higher (upwards of 50%) than in non-carriers.12,13 It is becoming exceedingly important to identify women who carry BRCA mutations so that they may utilize the latest medical advances in prevention, early detection, and treatment.3,14–19 To facilitate identification and referral of patients most likely to benefit from these advances, the National Comprehensive Cancer Network (NCCN) publishes annual guidelines that outline referral criteria for genetic counseling and testing in the oncology care setting.20 NCCN criteria for appropriate referrals in those with a personal and family history of breast cancer include, but are not limited to, early age of diagnosis of breast cancer (ie, ≤ age 50), number of relatives with breast cancer, two breast cancer primaries, triple-negative disease, male breast cancer, and previously identified BRCA mutation in the family (Table 1). Other considerations for referral include ethnic background, limited family size/structure, and presence of other BRCA-associated cancers in the family.
Table 1.
NCCN Referral Criteria for Further Genetic Risk Evaluation for HBOC
An affected individual with one or more of the following:
|
An unaffected individual with a family history of one or more of the following:
|
Reproduced/adapted with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Genetic/Familial High-Risk Assessment: Breast and Ovarian. V.1.2012. © 2012 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines, go online to www.NCCN.org. NATIONAL COMPREHENSIVE CANCER NETWORK®, NCCN®, NCCN GUIDELINES™, and all other NCCN Content are trademarks owned by the National Comprehensive Cancer Network, Inc.
Several models have been developed that predict the probability of a BRCA mutation in individuals or families, including those based on Bayesian approaches and statistical models21 or empiric observations (such as those based on Myriad prevalence tables).22 Overall, the use of prediction models has been shown to improve the ability of clinicians as to which patients are most likely to harbor a BRCA mutation.23 Studies that compared the different models suggest that no single model is consistently superior to others.24,25 Additional means shown to better improve the performance of prediction models and discriminate between BRCA1 and BRCA2 are inclusion of breast tumor markers such the estrogen receptor, progesterone receptor, and Her-2/neu status.26–28
Prevention
A number of studies have evaluated medical and surgical options for breast cancer prevention in BRCA mutation carriers. Tamoxifen has been the primary agent evaluated in the medical context. However, its role for primary prevention in BRCA carriers remains unclear. Specifically, a single small study of 8 BRCA1 carriers and 11 BRCA2 carriers showed a 62% reduction in breast cancer incidence in BRCA2 carriers who used tamoxifen, but no similar risk reduction was observed in BRCA1 carriers. To date, no larger studies have been conducted to examine the efficacy of tamoxifen for primary prevention of breast cancer. Additionally, there has been speculation that poly ADP-ribose polymerase (PARP) inhibitors may someday be considered as targeted chemoprevention agents for primary prevention of breast cancers in BRCA mutation carriers,29 and future efficacy studies are expected. For women with a prior breast cancer diagnosis, several studies have demonstrated a 50% to 70% reduction in risk for contralateral disease with use of tamoxifen.30–32 While promising, each of these studies have limitations such as retrospective design and lack of histopathological data (especially estrogen and progesterone receptor status) of the primary tumor.
In terms of surgical options, bilateral prophylactic mastectomy represents the highest level of risk reduction in BRCA mutation carriers. Multiple studies demonstrate a greater than 90% reduction in breast cancer risk.33–39 (For an overview of bilateral mastectomy in the context of a breast cancer diagnosis, refer to the Treatment section.) Another surgical option for breast cancer prevention is prophylactic bilateral salpingo-oophorectomy, which reduces ovarian cancer risk by approximately 80%40,41 and also confers a 50% reduction in breast cancer risk when performed premenopausally in BRCA carriers.41–43 It is unclear whether the magnitude of breast cancer risk reduction depends on time since oophorectomy, particularly when it is performed premenopausally, although available data suggest prophylactic oophorectomy performed after the age of 50 likely does not lower breast cancer risk.44 In terms of survival, prophylactic bilateral salpingo-oophorectomy has been reported to lead to a reduction in breast and ovarian cancer-specific mortality as well as overall mortality.39 Finally, there are data to suggest that prophylactic oophorectomy may also reduce contralateral breast cancer risk.45 A recent computer-simulated survival analysis examined the impact of screening and prophylactic surgeries in BRCA mutation carriers46 and developed an online tool for guiding management in patients.47 Recognizing the numerous assumptions on which modeling studies are based, these studies may nevertheless provide additional information for both patients and providers when making challenging management decisions.
Early Detection and Diagnosis
Several studies in BRCA mutation carriers have evaluated the impact of various breast cancer screening modalities on early cancer detection. For women with a BRCA mutation, the NCCN has issued recommendations for breast cancer surveillance,48 including monthly breast self-examination, semiannual clinical breast examination, annual mammogram, and annual breast magnetic resonance imaging (MRI), as illustrated in Table 2. Although yearly mammograms starting as early as 25 years of age are recommended as part of the screening strategy in BRCA mutation carriers, one potential concern is that BRCA carriers may be more prone to radiation-induced breast cancers than nonmutation carriers due to the role of the BRCA genes in DNA repair.49 However, there is currently insufficient evidence to indicate that radiation exposure from mammograms increases breast cancer risk.50–52
Table 2.
NCCN Management Recommendations for HBOC Syndrome Management
Women
|
Risk to Relatives
|
Reproductive Options
|
Reproduced/adapted with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Genetic/Familial High-Risk Assessment: Breast and Ovarian. V.1.2012. © 2012 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and illustrations herein may not be reproduced in any form for any purpose without the express written permission of the NCCN. To view the most recent and complete version of the NCCN Guidelines, go online to www.NCCN.org. NATIONAL COMPREHENSIVE CANCER NETWORK®, NCCN®, NCCN GUIDELINES™, and all other NCCN Content are trademarks owned by the National Comprehensive Cancer Network, Inc.
Studies have consistently shown that MRI is more sensitive than mammography and/or breast ultrasound in detection of early-stage breast cancers in BRCA mutation carriers.53–58 Furthermore, a prospective study of BRCA mutation carriers annually screened by MRI reported a significant reduction in the incidence of advanced breast cancer compared with those not screened by MRI.59 The optimal benefit of MRI appears to be achieved through annual screening between the ages of 25 to 65 years. The benefits of screening in this age group likely do not persist much beyond 2 years after the date of the last MRI; moreover, the long-term health implications of 40 doses of gadolinium in BRCA carriers have not been evaluated.44 Although trials indicate that MRI is superior to mammography in the detection of breast cancer, mammography identifies some cancers not detected by MRI, particularly ductal carcinoma in situ with microcalcifications.60,61 As a consequence, current evidence supports the benefits of both mammography and breast MRI annually in BRCA mutation carriers.3,46,62–64 Despite the proven benefits of MRI for breast cancer detection53–58 and recent data which suggest that it reduces the risk of advanced breast cancer,59 it is currently unknown if MRI results in a survival benefit.
Treatment
Several treatment considerations should be addressed for women with breast cancer who carry a BRCA mutation, including the high risks for contralateral disease and ovarian cancer, as well as the emergence of targeted treatment approaches currently offered in the context of clinical trials. Contralateral breast cancer rates in BRCA carriers are higher than that of non-carriers, with 10-year rates reported as 25% to 27% for carriers and 1% to 9% for non-carriers.65 Furthermore, recent studies indicate that contralateral breast cancer risk depends of age at first breast cancer diagnosis (with substantially elevated risks in those diagnosed at an early age) as well as affected BRCA gene (with higher risks for those with BRCA1 mutations).12,13 In fact, the lifetime contralateral breast cancer risk was reported to be over 60% in those with BRCA1 mutations diagnosed below the age of 40 after 25 years of follow-up.12 Because the presence of a germline BRCA mutation diagnosed at the time of a breast cancer diagnosis raises an individual’s risk of a synchronous and metachronous tumor, surgical treatment decision is often influenced by this knowledge. Due to this increased risk of a second primary breast cancer, approximately half of North American women with a BRCA mutation who are diagnosed with unilateral breast cancer select bilateral mastectomy for surgical treatment.66 This finding is supported by two US-based studies of newly diagnosed high-risk breast cancer patients who underwent BRCA testing at or near the time of diagnosis. In both studies, women who tested positive for a BRCA mutation chose bilateral mastectomy more often than women who received negative or uninformative results.67,68
A number of organizations provide guidelines to identify, refer, and counsel newly diagnosed high-risk breast cancer patients for genetic counseling, including those from the American Society of Clinical Oncology (originally published in 2003)69–71 and the American Society of Breast Surgeons (issued in 2009).72 However, a 2011 study of administrative data from a sample of 14.4 million commercially insured patients over a 3-year period found that of approximately 1,500 women with early-onset breast cancer ≤ age 40, only 30% had genetic testing, with even lower rates among black and Hispanic patients.73 Importantly, this study found that even among those who had testing (and presumably pretest genetic counseling), less than 20% did so within a month of diagnosis, suggesting that testing (and presumably pretest genetic counseling) generally occurred after definitive surgical treatment.
When considering ipsilateral breast cancer risk, a number of studies support breast conservation therapy (ie, lumpectomy and radiation) for treatment of the primary tumor, based on similar ipsilateral recurrence rates in carriers and non-carriers at 10 years of follow-up.12,74–76 However, at 15 years of follow-up, the risk of ipsilateral events (including second primary cancer and recurrence of initial cancer) has been reported to be as high as 24%.74,76 Ultimately, there is no documented difference in overall survival of BRCA carriers choosing breast conservation therapy vs mastectomy at 15 years following treatment.74 However, some data suggest it may take 20 years or more of follow-up to definitively determine whether bilateral mastectomy results in a mortality reduction.44,77 Another consideration for BRCA mutation carriers who select lumpectomy and radiation therapy but who may consider bilateral mastectomy in the future is the potential impact of radiation on breast reconstruction options. Changes to the skin caused by radiation have been well documented,78 and several reports have substantiated that implant reconstructions subsequent to radiation therapy have a significantly higher complication rate.79 Given the possibility of fewer options, BRCA mutation carriers should be fully informed about the impact of radiation therapy on future breast reconstruction.
While the influence of BRCA mutation carrier status on surgical decisions has become more widely considered in treatment planning,80 options to inform medical treatment strategies based on BRCA mutation carrier status are imminent in the era of personalized medicine. For example, preclinical models suggest BRCA1-associated breast cancers are resistant to taxanes and sensitive to DNA damaging agents such as mitomycin C and cisplatin.44 Subsequently, data have emerged to suggest that taxanes may not be useful in neoadjuvant treatment of BRCA1-associated breast cancers,81–84 whereas these cancers may be particularly sensitive to platinum agents.85,86 Currently, it remains unclear whether responsiveness to platinum-based agents in BRCA1-related breast cancers relates to the triple-negative phenotype or specifically to the BRCA mutation status itself. Interestingly, in a study of 28 women with stage II/III triple-negative breast cancer treated with cisplatin, 6 patients achieved complete clinical response, which included 2 women with BRCA1 mutations in the entire sample.87 However, more data are needed regarding the use of cisplatin in the clinical setting before clinical recommendations can be made.
Among the many tailored treatments currently under investigation,88 much interest has focused on the PARP inhibitors, which may be particularly effective in BRCA-associated cancers.89–92 This is the first treatment regimen to be based on a synthetic lethality approach, as inhibiting PARP-mediated pathways selectively affects only those cells that have lost BRCA-associated homologous recombination function (ie, tumor cells), whereas normal cells are unaffected. Consequently, this approach is associated with minimal systemic toxicity.18 Several international trials are underway to test the effectiveness of PARP inhibitors in BRCA mutation carriers,17–19,91,93–95 and many have reported promising preliminary results.17–19 Notably, single-agent PARP (olaparib) has shown effectiveness in treating metastatic breast cancer.18,96
In individuals with a new diagnosis of breast cancer and determined to have a BRCA mutation, another concern is the high risk of metachronous ovarian cancer, which is a common cause of mortality in those BRCA mutation carriers who were successfully treated for early-stage breast cancers.97 Specifically, patients with a BRCA mutation also have a 7% to 13% chance of developing ovarian cancer within 10 years after diagnosis.97 As surveillance has not been shown to be effective in detecting early-stage ovarian cancers, the benefits of bilateral salpingo-oophorectomy (in addition to lowering the risk of breast cancer) are related to their effect on survival from ovarian cancer.97 Further discussion of bilateral salpingo-oophorectomy is included below (related to the management of symptoms associated with menopause) and in the Prevention section (related to breast cancer risk reduction).
Quality of Life and Survivorship
Several important considerations regarding quality of life and survivorship are unique to women who carry a BRCA mutation, including the impact of risk-reducing surgery, fertility and reproductive health concerns, communication of genetic test results, and concerns about insurability and discrimination. Overall, the published literature indicates high levels of satisfaction (80% to 100%) with prophylactic mastectomy over time.98–102 One of the first studies of psychosocial outcomes of prophylactic mastectomy to include only women who were genetically susceptible to breast cancer was published in 2006.103 Among the 114 Dutch women who underwent prophylactic mastectomy and breast reconstruction, after a median follow-up of 3 years, 60% of participants were satisfied with the results of the surgery. The sample included 63 women with BRCA mutations and 14 at high risk for breast cancer by virtue of their strong family history. Those who perceived a lack of information, experienced complications, had ongoing feelings that the reconstructed breasts did not feel like their own, and stated they would not choose this type of reconstruction again were least likely to be satisfied and to report adverse effects on their sexual relationship. Additionally, negative impacts on the patient’s sexual relationship were also associated with altered feelings of femininity, discrepant expectations, and perceptions that the partner had a negative perception of femininity and sexuality. The level of satisfaction reported in this study is lower than what has previously been reported in women who were not specifically identified as BRCA mutation carriers. Since this study was conducted in the Netherlands, there may be some variation specific to the healthcare system; however, it is also possible that there are psychosocial issues specific to BRCA mutation-positive women undergoing prophylactic mastectomy that warrant further exploration.
For women who undergo bilateral prophylactic oophorectomy, evidence suggests an improvement in psychosocial functioning after surgery. Among 846 Dutch women at high risk for HBOC, 44% selected prophylactic bilateral salpingo oophorectomy and 56% selected surveillance.104 Women who selected oophorectomy had fewer breast and ovarian cancer worries compared with those selecting surveillance. However, those who selected surgery were also significantly more likely to report menopausal symptoms and worse sexual functioning (eg, less pleasure during sexual activities). The use of hormone replacement therapy (HRT) to mitigate the symptoms associated with surgical menopause in young women who undergo bilateral prophylactic oophorectomy has been an issue of debate. A concern about HRT in BRCA carriers is its potential to raise the risk of breast cancer, as seen in the general population.105–107 However, two studies in BRCA mutation carriers reported that HRT did not increase the subsequent risk of breast cancer, nor did it appear to reduce the protective effect of oophorectomy on breast cancer risk.108,109 Recent results from the Women’s Health Initiative in the general population also provide reassurance as to estrogen use for about 5 years in terms of breast cancer risk and mortality.110 Ultimately, HRT mitigates many of the symptoms associated with menopause. However, studies have suggested that quality of life does not return to presurgical levels.104,111
Fertility has been identified as a leading quality of life concern among breast cancer patients diagnosed during their reproductive years.112 Estimated infertility risks for women treated with adjuvant breast cancer therapy for 6 weeks with the most commonly used chemotherapy regimens are > 80% for premenopausal women aged 40 and above, 20% to 80% for women aged 30 to 39, and < 20% for women under age 30.113 In recognition of the immediacy of fertility concerns, professional guidelines have been released by both the American Society for Reproductive Medicine (ASRM) in 2005 and the American Society of Clinical Oncology (ASCO) in 2006 that highlight the role of oncologists to initiate discussion of fertility so their patients can make family-building decisions that are right for them.113,114 Despite these guidelines, research suggests a lack of patient knowledge and a need for improved communication about fertility between cancer patients and their providers. A survey of cancer survivors found that only 57% of patients report receiving information about fertility from healthcare providers.115 Another study of young female cancer survivors reported a higher discussion rate of 72%.116 However, only 51% of these women felt their concerns were addressed adequately, suggesting that even if this communication is occurring, patients may not receive sufficient information regarding fertility risk and fertility preservation options. Finally, a national study of US oncologists found that 47% of respondents reported routinely referring cancer patients of childbearing age to a reproductive endocrinologist.117
The possibility of transmitting a mutation to a child may also pose a concern to families with a BRCA mutation,118 perhaps to the extent that some carriers may avoid childbearing.119,120 These concerns may prompt women to consider using a preimplantation genetic diagnosis (PGD), a newer technique used to test fertilized embryos for genetic disorders prior to uterine implantation.121–124 PGD involves the use of in vitro fertilization, in which an egg is fertilized and develops for 3 days until it reaches the 8-cell stage, at which time 1 cell is removed and examined using polymerase chain reaction (PCR) or fluorescence in situ hybridization (FISH).122 Using the information obtained from the genetic testing, potential parents can decide whether or not to implant embryos. A review by Offit et al125 indicated that PGD has been used in the context of several hereditary cancer syndromes, including HBOC. Given the immediate and long-term issues related to reproductive health based on medical and surgical treatment and risk reduction options for BRCA mutation carriers, the optimal time for oncology care providers to discuss fertility-related concerns is prior to the initiation of chemotherapy.126
A primary motivation for many breast cancer patients to undergo BRCA testing is to provide information to their at-risk relatives.127–130 In clinical practice, healthcare providers give test results directly to patients and encourage them to share the results with at-risk family members.131 Several studies to date have documented that women generally share BRCA test results with their at-risk relatives.132 However, some studies indicate that females who have been tested may choose less frequently to share results with their relatives who are male, with those they do not feel emotionally close to, or with those whose emotional or psychological well being is a concern.133–137 Although likely uncommon, a recent case report documented deliberate disclosure of incorrect test results by a patient to her at-risk family member.138 There is also recent literature suggesting that parents are disclosing test results to minor children.139–141 While there is no specific clinical guidelines about the appropriate age to disclose a parent’s test results to children, there are clear guidelines against testing minor children for BRCA mutations. Consensus guidelines from experts consistently emphasize that genetic testing for adult onset conditions should be deferred until legal adulthood (age 18), when individuals can decide for themselves whether they would like to pursue testing.70,142–144 This guideline reflects concerns regarding autonomy, potential discrimination, and possible psychological effects.142,144,145 Therefore, in HBOC, where there is no risk of malignancy in childhood and no risk reduction strategies are recommended in minors, testing should not be offered. Even after 18 years of age, testing is generally not strongly recommended due to potential implications to future insurability in the absence of any medical interventions implemented at this age. The age at which screening strategies are initiated is in the mid 20s (Table 2), which is typically the time at which many clinicians recommend testing for at risk women.
Other concerns expressed by women considering BRCA testing are those related to genetic discrimination and insurability. Specifically, when embarking on genetic testing for inherited cancer susceptibility, particularly in the context of a family member who does not have a cancer diagnosis, potential implications regarding future insurability are an important aspect of discussion during the genetic counseling session. At the federal level, the Genetic Information Non-Discrimination Act (GINA) was passed in 2008, which imparts protections for individual and group health insurance policies as well as in the workplace.146 Note that GINA does not cover life insurance, disability insurance, or other supplemental insurances. In addition, those in the military are not covered under GINA. However, other protections against discrimination are in place.147
As patients move from treatment to survivorship, it is important to consider their transition back to primary care providers. Given the strict laws in some states with regard to confidentiality of genetic test results, some centers limit documentation of BRCA mutation results in the medical record and/or implement additional safety guards to house and release this information within the medical record. Consequently, patients may share some responsibility to inform their healthcare providers of their mutation results, particularly as they are transitioning back to primary care settings. This information will be useful for providers to not only make cancer screening recommendations based on mutation status, but also ensure that patients follow recommended screening and behavioral guidelines for prevention and early detection of non–BRCA-related cancers.148
These quality of life and survivorship issues in conjunction with the difficulty some patients may have to accurately recall their test results149–151 underscore the importance of pre- and posttest genetic counseling that is provided by a knowledgeable genetics professional. In addition to expertise in cancer risk assessment and medical management of individuals with hereditary cancer predisposition, these individuals are also trained to identify potential psychosocial and educational needs of patients.152 They are critical members of the healthcare team and can help ensure that patients understand the medical and psychosocial implications of results for themselves and at-risk family members and serve as a resource for patients to develop strategies for effective communication.
Considerations for Delivery of BRCA Counseling and Testing in the Breast Oncology Care Setting
Several health and professional organizations strongly encourage referral to a certified/credentialed cancer genetics professional for pretest counseling, prior to genetic testing.20,70,143,152–156 There are three main groups of healthcare professionals with advanced degrees and experience in genetics. (1) A Certified Genetic Counselor (CGC) has at least a Master’s degree in genetic counseling and has passed a national board examination, (2) a Diplomate, American Board of Medical Genetics (DABMG), or Fellow, American College of Medical Genetics (FACMG) is a physician who has completed residency or fellowship training in medical genetics and passed the board examination, and (3) an Advanced Practice Nurse in Genetics (APNG) has completed both a graduate nursing program and professional portfolio review process.157 As outlined in a recent publication,158 the traditional model of delivering genetic counseling and testing services involve five steps: patient identification, genetic consultation, genetic testing if appropriate, posttest counseling, and cancer risk management.
Patients are referred by healthcare providers or family members, or they are self-referred. The model for providing comprehensive BRCA testing begins with an in-person pretest genetic counseling session during which time informed consent for genetic testing is obtained followed by a results disclosure session.16 During this session, the patient typically meets with a genetics professional with the training described above. The professional obtains personal and family cancer history and provides the patient with a risk assessment, including which, if any, genetic tests may be appropriate to perform. Given the complexities of delivering high-quality genetics services, ASCO provides key points that should be discussed with any patient during a pretest genetic counseling session (Table 3).70,143 This initial appointment typically lasts 60 to 90 minutes. One of the primary goals of the initial session is to determine whether genetic testing is appropriate for a given patient and then proceed accordingly.152 For those who proceed with testing, the patient returns for in-person disclosure of test results and discussion of medical management options. However, over the last decade, the delivery of genetics services (including who provides these services) has evolved, resulting in new models to optimize development of and access to these services.158–160 Increasingly, comprehensive services for hereditary breast cancer are delivered by a multidisciplinary team that includes genetic counselors working in conjunction with oncologists, medical geneticists, other medical specialists, and often a mental health professional.161 This interdisciplinary approach can help avoid problems such as inappropriate or incomplete testing, misinterpretation of test results by both patients and clinicians, inappropriate cancer screening and prevention recommendations, and psychological issues.71,162,163
Table 3.
Basic Elements of Informed Consent for Cancer Susceptibility Testing
|
From American Society of Clinical Oncology. American Society of Clinical Oncology policy statement update: genetic testing for cancer susceptibility. J Clin Oncol. 2003;21(12):2397–2406. Reprinted with permission. © 2003 American Society of Clinical Oncology. All rights reserved.
Compared with most other clinical services, provision of genetic counseling involves significant amounts of provider time.160,164–167 However, low reimbursement rates relative to the time required to provide adequate services impedes the provision of adequate risk counseling, particularly for providers outside of an academic setting.168 One study found that only one-third of the time needed to deliver genetic counseling services involves face-to-face patient care that can be billed.169 Specifically, most genetic counseling programs cost more than the direct revenue they generate.77 However, another crucial consideration to illustrate institutional value of genetic counseling programs must incorporate “downstream revenue analysis,” which includes reimbursement from surveillance, prophylactic surgeries, or cancer treatments for individuals with BRCA mutations.170 Thus, the inclusion of this income is essential in any cost-benefit calculation of genetic counseling services, and additional research regarding this revenue is needed. Ultimately, public and private healthcare policy reform is needed to address the gap in insurance coverage for adequate risk counseling and genetic analyses as a component of preventive care.
Institutional Efforts Specific to Genetic Counseling and Testing
At our institute, a model driven by physician recommendation has been implemented as a referral system for recently diagnosed high-risk breast cancer patients. The available personal and family history records for newly diagnosed breast cancer patient are reviewed by a genetic counselor during the weekly Breast Program tumor board meetings to assess if the patient meets NCCN guidelines for referral to the Cancer Genetic Counseling and Testing Service, as illustrated in Table 1.20 Each patient who is discussed and meets these criteria is noted by the genetic counselor. A letter is generated by the Cancer Genetic Counseling and Testing Service and sent to the surgeon for his or her signature. In addition to a surgeon discussing genetic counseling and testing during the initial consultation, a referral letter signed by the patient’s physician (often a surgeon) is sent to a breast cancer patient who has been identified as meeting NCCN criteria. This brief letter is addressed directly to the patient, signed by his or her surgeon, and recommends that the patient make an appointment for genetic counseling.171 In an effort to continually improve our delivery of clinical services, our team evaluates important outcomes such as patient uptake of and satisfaction with genetics services.172,173 These data are in turn used to generate additional research opportunities. For example, we were recently awarded peer-reviewed funding to develop and test an educational intervention specific to postsurgical breast cancer patients at risk for HBOC who are transitioning to survivorship care.
While the physician referral model works well in settings with access to trained genetics professionals, there are many rural areas or certain states where such professionals are not readily available.174,175 This shortage is compounded by the paucity of education and training opportunities in clinical cancer genetics,176 despite priorities set forth by key stakeholders emphasizing the need for cancer genetics education.177–185 Thus, integrating genetic counseling and testing for HBOC into clinical care remains an ongoing challenge. However, several innovative approaches to delivering quality genetics services to an increasing number of patients in community settings have been developed. For example, successful strategies for delivering comprehensive genetic counseling and testing services include academic-community partnerships that focus on collaboration with nongenetics providers to offer genetic testing for hereditary cancers.158,159 This approach can leverage the expertise of an academic center for challenging cases as well as allow patients to remain in their community and provide better access to resources for long-term follow-up care. Other innovative approaches to successfully identify high-risk patients, particularly in the primary care and community setting, include automated family collection tools through which risk assessment is performed at a hospital or practice-wide level and appropriate patients are identified and referred for genetic counseling and testing.186–188 These software tools are particularly of interest as many hospitals and practices transition to electronic health records, and they have the potential to enhance the identification of patients at risk for inherited cancers at a system-wide level.
At our institute, we secured peer-reviewed funding to develop an infrastructure (called the Inherited Cancer Research [ICARE] Initiative) to support research, education, and outreach initiatives focused on BRCA genetic counseling and testing. The ICARE Initiative leverages a state mandate to reach the citizens of Florida and provide access to high-quality cancer care. Moffitt works with its partners (referred to as “affiliates”) to offer clinical expertise and research trials found at an NCI-designated Comprehensive Cancer Center. The Moffitt Affiliate Network (MAN) Program represents strategic affiliations with 15 Florida hospitals, 1 Georgia hospital, and more than 400 community oncologists, through which Moffitt reaches approximately 20% of Florida cancer patients. Through networking with our MAN sites, providing access to a genetic counselor for general questions, and utilizing other directed learning opportunities and educational resources/materials over the last year, 12 of the 15 sites have actively participated in our bimonthly case conferences. Furthermore, an additional 2 sites have requested and received in-person presentations from the ICARE team on the topic of inherited cancer susceptibility. In addition to our educational and outreach efforts, MAN practitioners refer high-risk patients to our research registry to provide the research link, which has in turn contributed to the tremendous growth of our registry since initiation of the grant in the summer of 2010. This provides unprecedented opportunities to understand cancer risk management practices and recommendations in BRCA mutation carriers at the patient and practitioner level. These efforts will provide information that can improve the delivery of cancer genetics services in the state of Florida and serve as a model for other states.
Conclusions
Over the last several years, it has become increasingly common to consider BRCA mutation status when determining optimal cancer risk management and treatment options in order to improve patient outcomes. Knowledge about the risk for hereditary cancer at or as close as possible to the time of diagnosis allows patients access to options that may improve their outcome and reduce treatment-associated risks. Thus it has become important for providers in the breast oncology care setting to identify at-risk patients and offer testing, either by themselves or through referral to a cancer genetic counseling and testing service. As such, leading health professional organizations such as the National Comprehensive Cancer Network,20 the American Society of Breast Surgeons,72 and the American Society of Clinical Oncology189 provide specific guidelines and recommendations to providers for identification of women at increased risk for carrying a BRCA mutation. For those providers without in-house clinical cancer genetics providers, several promising collaborative models across the United States are successfully bringing the expertise of these providers to breast oncology care for patients in community settings.158,159
Acknowledgments
This work was supported in part by grants through Florida Biomedical (IBG09-34198) and the National Human Genome Research Institute (R21HG006415).
Footnotes
No significant relationship exists between the authors and the companies/organizations whose products or services may be referenced in this article.
Appreciation is expressed to Veronica Harville for administrative assistance.
References
- 1.Claus EB, Schildkraut JM, Thompson WD, et al. The genetic attributable risk of breast and ovarian cancer. Cancer. 1996;77(11):2318–2324. doi: 10.1002/(SICI)1097-0142(19960601)77:11<2318::AID-CNCR21>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
- 2.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. 2001;68(3):700–710. doi: 10.1086/318787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Narod SA, Offit K. Prevention and management of hereditary breast cancer. J Clin Oncol. 2005;23(8):1656–1663. doi: 10.1200/JCO.2005.10.035. [DOI] [PubMed] [Google Scholar]
- 4.Malone KE, Daling JR, Doody DR, et al. Prevalence and predictors of BRCA1 and BRCA2 mutations in a population-based study of breast cancer in white and black American women ages 35 to 64 years. Cancer Res. 2006;66(16):8297–8308. doi: 10.1158/0008-5472.CAN-06-0503. [DOI] [PubMed] [Google Scholar]
- 5.Pal T, Permuth-Wey J, Betts JA, et al. BRCA1 and BRCA2 mutations account for a large proportion of ovarian carcinoma cases. Cancer. 2005;104(12):2807–2816. doi: 10.1002/cncr.21536. [DOI] [PubMed] [Google Scholar]
- 6.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. 1998;178(4):670–677. doi: 10.1016/s0002-9378(98)70476-4. [DOI] [PubMed] [Google Scholar]
- 7.Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet. 2003;72(5):1117–1130. doi: 10.1086/375033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Litton JK, Ready K, Chen H, et al. Earlier age of onset of BRCA mutation-related cancers in subsequent generations. Cancer. 2012;118(2):321–325. doi: 10.1002/cncr.26284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol. 2007;25(11):1329–1333. doi: 10.1200/JCO.2006.09.1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.King MC, Marks JH, Mandell JB. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science. 2003;302(5645):643–646. doi: 10.1126/science.1088759. [DOI] [PubMed] [Google Scholar]
- 11.Moller P, Maehle L, Vabo A, Clark N, Sun P, Narod S. Age-specific incidence rates for breast cancer in carriers of BRCA1 mutations from Norway. Clin Genet. Feb 9; doi: 10.1111/j.1399-0004.2012.01855.x. [DOI] [PubMed] [Google Scholar]
- 12.Graeser MK, Engel C, Rhiem K, et al. Contralateral breast cancer risk in BRCA1 and BRCA2 mutation carriers. J Clin Oncol. 2009;27(35):5887–5892. doi: 10.1200/JCO.2008.19.9430. [DOI] [PubMed] [Google Scholar]
- 13.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. 2010;28(14):2404–2410. doi: 10.1200/JCO.2009.24.2495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Watson M, Kash KM, Homewood J, et al. Does genetic counseling have any impact on management of breast cancer risk? Genet Test. 2005;9(2):167–174. doi: 10.1089/gte.2005.9.167. [DOI] [PubMed] [Google Scholar]
- 15.Roukos DH, Briasoulis E. Individualized preventive and therapeutic management of hereditary breast ovarian cancer syndrome. Nat Clin Pract Oncol. 2007;4(10):578–590. doi: 10.1038/ncponc0930. [DOI] [PubMed] [Google Scholar]
- 16.Weitzel JN, Blazer KR, Macdonald DJ, et al. Genetics, genomics, and cancer risk assessment: state of the art and future directions in the era of personalized medicine. CA Cancer J Clin. 2011 Aug 19; doi: 10.3322/caac.20128. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Audeh MW, Penson RT, Friedlander M, et al. Phase II trial of the oral PARP inhibitor olaparib (AZD2281) in BRCA-deficient advanced ovarian cancer. J Clin Oncol. 2009;27:15s. (suppl; abstr 5500) [Google Scholar]
- 18.Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361(2):123–134. doi: 10.1056/NEJMoa0900212. [DOI] [PubMed] [Google Scholar]
- 19.Tutt A, Robson M, Garber JE, et al. Phase II trial of the oral PARP inhibitor olaparib in BRCA-deficient advanced breast cancer. J Clin Oncol. 2009;27(15 suppl):CRA501. Abstract. [Google Scholar]
- 20.Genetic/Familial High-Risk Assessment: Breast and Ovarian. [Accessed June 21, 2012];NCCN Practice Guidelines. V.1.2012 http://www.nccn.org/professionals/physician_gls/pdf/genetics_screening.pdf.
- 21.Parmigiani G, Berry D, Aguilar O. Determining carrier probabilities for breast cancer-susceptibility genes BRCA1 and BRCA2. Am J Hum Genet. 1998;62 (1):145–158. doi: 10.1086/301670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Myriad Genetics, Laboratories and Pharmaceuticals. [Accessed June 21, 2012]; http://www.myriadtests.com/provider/brca-mutation-prevalence.htm.
- 23.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. 2002;94(11):844–851. doi: 10.1093/jnci/94.11.844. [DOI] [PubMed] [Google Scholar]
- 24.Kang HH, Williams R, Leary J, et al. Evaluation of models to predict BRCA germline mutations. Br J Cancer. 2006;95(7):914–920. doi: 10.1038/sj.bjc.6603358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Antoniou AC, Hardy R, Walker L, et al. Predicting the likelihood of carrying a BRCA1 or BRCA2 mutation: validation of BOADICEA, BRCAPRO, IBIS, Myriad and the Manchester scoring system using data from UK genetics clinics. J Med Genet. 2008;45(7):425–431. doi: 10.1136/jmg.2007.056556. [DOI] [PubMed] [Google Scholar]
- 26.Tai YC, Chen S, Parmigiani G, et al. Incorporating tumor immunohistochemical markers in BRCA1 and BRCA2 carrier prediction. Breast Cancer Res. 2008;10(2):401. doi: 10.1186/bcr1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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. 2012;133(1):347–355. doi: 10.1007/s10549-012-1958-z. [DOI] [PubMed] [Google Scholar]
- 28.Mavaddat N, Rebbeck TR, Lakhani SR, et al. Incorporating tumour pathology information into breast cancer risk prediction algorithms. Breast Cancer Res. 2010;12(3):R28. doi: 10.1186/bcr2576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rubinstein WS. Hereditary breast cancer: pathobiology, clinical translation, and potential for targeted cancer therapeutics. Fam Cancer. 2008;7(1):83–89. doi: 10.1007/s10689-007-9147-7. [DOI] [PubMed] [Google Scholar]
- 30.Gronwald J, Tung N, Foulkes WD, et al. Tamoxifen and contralateral breast cancer in BRCA1 and BRCA2 carriers: an update. Int J Cancer. 2006;118(9):2281–2284. doi: 10.1002/ijc.21536. [DOI] [PubMed] [Google Scholar]
- 31.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. 2000;356(9245):1876–1881. doi: 10.1016/s0140-6736(00)03258-x. [DOI] [PubMed] [Google Scholar]
- 32.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. 2006;24(16):2437–2443. doi: 10.1200/JCO.2005.02.7888. [DOI] [PubMed] [Google Scholar]
- 33.Hartmann LC, Sellers TA, Schaid DJ, et al. Efficacy of bilateral prophylactic mastectomy in BRCA1 and BRCA2 gene mutation carriers. J Natl Cancer Inst. 2001;93(21):1633–1637. doi: 10.1093/jnci/93.21.1633. [DOI] [PubMed] [Google Scholar]
- 34.Rebbeck TR, Friebel T, Lynch HT, et al. Bilateral prophylactic mastectomy reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE Study Group. J Clin Oncol. 2004;22(6):1055–1062. doi: 10.1200/JCO.2004.04.188. [DOI] [PubMed] [Google Scholar]
- 35.Meijers-Heijboer H, van Geel B, van Putten WL, et al. Breast cancer after prophylactic bilateral mastectomy in women with a BRCA1 or BRCA2 mutation. N Engl J Med. 2001;345(3):159–164. doi: 10.1056/NEJM200107193450301. [DOI] [PubMed] [Google Scholar]
- 36.van Sprundel TC, Schmidt MK, Rookus MA, et al. Risk reduction of contra-lateral breast cancer and survival after contralateral prophylactic mastectomy in BRCA1 or BRCA2 mutation carriers. Br J Cancer. 2005;93(3):287–292. doi: 10.1038/sj.bjc.6602703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Evans DG, Baildam AD, Anderson E, et al. Risk reducing mastectomy: outcomes in 10 European centres. J Med Genet. 2009;46(4):254–258. doi: 10.1136/jmg.2008.062232. [DOI] [PubMed] [Google Scholar]
- 38.Heemskerk-Gerritsen BA, Brekelmans CT, Menke-Pluymers MB, et al. Prophylactic mastectomy in BRCA1/2 mutation carriers and women at risk of hereditary breast cancer: long-term experiences at the Rotterdam Family Cancer Clinic. Ann Surg Oncol. 2007;14(12):3335–3344. doi: 10.1245/s10434-007-9449-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA. 2010;304(9):967–975. doi: 10.1001/jama.2010.1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Finch A, Beiner M, Lubinski J, et al. Salpingo-oophorectomy and the risk of ovarian, fallopian tube, and peritoneal cancers in women with a BRCA1 or BRCA2 Mutation. JAMA. 2006;296(2):185–192. doi: 10.1001/jama.296.2.185. [DOI] [PubMed] [Google Scholar]
- 41.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. 2009;101(2):80–87. doi: 10.1093/jnci/djn442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Eisen A, Lubinski J, Klijn J, et al. Breast cancer risk following bilateral oophorectomy in BRCA1 and BRCA2 mutation carriers: an international case-control study. J Clin Oncol. 2005;23(30):7491–7496. doi: 10.1200/JCO.2004.00.7138. [DOI] [PubMed] [Google Scholar]
- 43.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. 2008;26(8):1331–1337. doi: 10.1200/JCO.2007.13.9626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Narod SA. BRCA mutations in the management of breast cancer: the state of the art. Nat Rev Clin Oncol. 2010;7(12):702–707. doi: 10.1038/nrclinonc.2010.166. [DOI] [PubMed] [Google Scholar]
- 45.Metcalfe K, Gershman S, Lynch HT, et al. Predictors of contralateral breast cancer in BRCA1 and BRCA2 mutation carriers. Br J Cancer. 2011;104(9):1384–1392. doi: 10.1038/bjc.2011.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kurian AW, Sigal BM, Plevritis SK. Survival analysis of cancer risk reduction strategies for BRCA1/2 mutation carriers. J Clin Oncol. 2010;28(2):222–231. doi: 10.1200/JCO.2009.22.7991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kurian AW, Munoz DF, Rust P, et al. Online tool to guide decisions for BRCA1/2 mutation carriers. J Clin Oncol. 2012;30(5):497–506. doi: 10.1200/JCO.2011.38.6060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bevers TB, Anderson BO, Bonaccio E, et al. NCCN clinical practice guidelines in oncology: breast cancer screening and diagnosis. J Natl Compr Canc Netw. 2009;7(10):1060–1096. doi: 10.6004/jnccn.2009.0070. [DOI] [PubMed] [Google Scholar]
- 49.Murphy CG, Moynahan ME. BRCA gene structure and function in tumor suppression: a repair-centric perspective. Cancer J. 2010;16(1):39–47. doi: 10.1097/PPO.0b013e3181cf0204. [DOI] [PubMed] [Google Scholar]
- 50.Narod SA, Lubinski J, Ghadirian P, et al. Screening mammography and risk of breast cancer in BRCA1 and BRCA2 mutation carriers: a case-control study. Lancet Oncol. 2006;7(5):402–406. doi: 10.1016/S1470-2045(06)70624-6. [DOI] [PubMed] [Google Scholar]
- 51.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-HE-BON, and IBCCS Collaborators’ Group. J Clin Oncol. 2006;24(21):3361–3366. doi: 10.1200/JCO.2005.03.3126. [DOI] [PubMed] [Google Scholar]
- 52.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. 2006;15(11):2311–2313. doi: 10.1158/1055-9965.EPI-06-0176. [DOI] [PubMed] [Google Scholar]
- 53.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. 2004;292(11):1317–1325. doi: 10.1001/jama.292.11.1317. [DOI] [PubMed] [Google Scholar]
- 54.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. 2005;365(9473):1769–1778. doi: 10.1016/S0140-6736(05)66481-1. [DOI] [PubMed] [Google Scholar]
- 55.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. 2010;28(9):1450–1457. doi: 10.1200/JCO.2009.23.0839. [DOI] [PubMed] [Google Scholar]
- 56.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. 2010;28(36):5265–5273. doi: 10.1200/JCO.2009.27.2294. [DOI] [PubMed] [Google Scholar]
- 57.Weinstein SP, Localio AR, Conant EF, et al. Multimodality screening of high-risk women: a prospective cohort study. J Clin Oncol. 2009;27(36):6124–6128. doi: 10.1200/JCO.2009.24.4277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sardanelli F, Podo F, Santoro F, et al. Multicenter surveillance of women at high genetic breast cancer risk using mammography, ultrasonography, and contrast-enhanced magnetic resonance imaging (the high breast cancer risk italian 1 study): final results. Invest Radiol. 2011;46(2):94–105. doi: 10.1097/RLI.0b013e3181f3fcdf. [DOI] [PubMed] [Google Scholar]
- 59.Warner E, Hill K, Causer P, et al. Prospective study of breast cancer incidence in women with a BRCA1 or BRCA2 mutation under surveillance with and without magnetic resonance imaging. J Clin Oncol. 2011;29(13):1664–1669. doi: 10.1200/JCO.2009.27.0835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.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. 2010;119(2):399–407. doi: 10.1007/s10549-009-0607-7. [DOI] [PubMed] [Google Scholar]
- 61.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. 2009;118(3):539–546. doi: 10.1007/s10549-009-0475-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gulati AP, Domchek SM. The clinical management of BRCA1 and BRCA2 mutation carriers. Curr Oncol Rep. 2008;10(1):47–53. doi: 10.1007/s11912-008-0008-9. [DOI] [PubMed] [Google Scholar]
- 63.Nusbaum R, Isaacs C. Management updates for women with a BRCA1 or BRCA2 mutation. Mol Diagn Ther. 2007;11(3):133–144. doi: 10.1007/BF03256234. [DOI] [PubMed] [Google Scholar]
- 64.Smith KL, Isaacs C. Management of women at increased risk for hereditary breast cancer. Breast Dis. 2006;27:51–67. doi: 10.3233/bd-2007-27104. [DOI] [PubMed] [Google Scholar]
- 65.Trainer AH, Lewis CR, Tucker K, et al. The role of BRCA mutation testing in determining breast cancer therapy. Nat Rev Clin Oncol. 2010;7(12):708–717. doi: 10.1038/nrclinonc.2010.175. [DOI] [PubMed] [Google Scholar]
- 66.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. 2008;122(9):2017–2022. doi: 10.1002/ijc.23340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Schwartz MD, Lerman C, Brogan B, et al. Impact of BRCA1/BRCA2 counseling and testing on newly diagnosed breast cancer patients. J Clin Oncol. 2004;22(10):1823–1829. doi: 10.1200/JCO.2004.04.086. [DOI] [PubMed] [Google Scholar]
- 68.Weitzel JN, McCaffrey SM, Nedelcu R, et al. Effect of genetic cancer risk assessment on surgical decisions at breast cancer diagnosis. Arch Surg. 2003;138(12):1323–1328. doi: 10.1001/archsurg.138.12.1323. discussion 1329. [DOI] [PubMed] [Google Scholar]
- 69.Statement of the American Society of Clinical Oncology: genetic testing for cancer susceptibility, Adopted on February 20, 1996. J Clin Oncol. 1996;14(5):1730–1736. doi: 10.1200/JCO.1996.14.5.1730. discussion 1737–1740. [DOI] [PubMed] [Google Scholar]
- 70.American Society of Clinical Oncology policy statement update: genetic testing for cancer susceptibility. J Clin Oncol. 2003;21(12):2397–2406. doi: 10.1200/JCO.2003.03.189. [DOI] [PubMed] [Google Scholar]
- 71.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. 2010;28(5):893–901. doi: 10.1200/JCO.2009.27.0660. [DOI] [PubMed] [Google Scholar]
- 72.The American Society of Breast Surgeons. [Accessed June 27, 2012];BRCA Genetic Testing for Patients With and Without Breast Cancer. 2006 http://www.breastsurgeons.org/statements/brca.php.
- 73.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. 2011;13(4):349–255. doi: 10.1097/GIM.0b013e3182091ba4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.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. 2010;121(2):389–398. doi: 10.1007/s10549-010-0894-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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. 2010;120(1):119–126. doi: 10.1007/s10549-009-0685-6. [DOI] [PubMed] [Google Scholar]
- 76.Metcalfe K, Lynch HT, Ghadirian P, et al. Risk of ipsilateral breast cancer in BRCA1 and BRCA2 mutation carriers. Breast Cancer Res Treat. 2011;127(1):287–296. doi: 10.1007/s10549-010-1336-7. [DOI] [PubMed] [Google Scholar]
- 77.Tung N. Management of women with BRCA mutations: a 41-year-old woman with a BRCA mutation and a recent history of breast cancer. JAMA. 2011;305(21):2211–2220. doi: 10.1001/jama.2011.678. [DOI] [PubMed] [Google Scholar]
- 78.Mansfield C. Effects of radiation therapy on wound healing after mastectomy. Clin Plast Surg. 1979;6(1):19–26. [PubMed] [Google Scholar]
- 79.Vandeweyer E, Deraemaecker R. Radiation therapy after immediate breast reconstruction with implants. Plast Reconstr Surg. 2000;106(1):56–58. doi: 10.1097/00006534-200007000-00009. discussion 59–60. [DOI] [PubMed] [Google Scholar]
- 80.Daly MB. Tailoring breast cancer treatment to genetic status: the challenges ahead. J Clin Oncol. 2004;22(10):1776–1777. doi: 10.1200/JCO.2004.02.994. [DOI] [PubMed] [Google Scholar]
- 81.Fedier A, Steiner RA, Schwarz VA, et al. The effect of loss of Brca1 on the sensitivity to anticancer agents in p53-deficient cells. Int J Oncol. 2003;22(5):1169–1173. [PubMed] [Google Scholar]
- 82.Chabalier C, Lamare C, Racca C, et al. BRCA1 downregulation leads to premature inactivation of spindle checkpoint and confers paclitaxel resistance. Cell Cycle. 2006;5(9):1001–1007. doi: 10.4161/cc.5.9.2726. [DOI] [PubMed] [Google Scholar]
- 83.Byrski T, Gronwald J, Huzarski T, et al. Response to neo-adjuvant chemotherapy in women with BRCA1-positive breast cancers. Breast Cancer Res Treat. 2008;108(2):289–296. doi: 10.1007/s10549-007-9600-1. [DOI] [PubMed] [Google Scholar]
- 84.Wysocki PJ, Korski K, Lamperska K, et al. Primary resistance to docetaxel-based chemotherapy in metastatic breast cancer patients correlates with a high frequency of BRCA1 mutations. Med Sci Monit. 2008;14(7):SC7–SC10. [PubMed] [Google Scholar]
- 85.Bhattacharyya A, Ear US, Koller BH, et al. The breast cancer susceptibility gene BRCA1 is required for subnuclear assembly of Rad51 and survival following treatment with the DNA cross-linking agent cisplatin. J Biol Chem. 2000;275(31):23899–23903. doi: 10.1074/jbc.C000276200. [DOI] [PubMed] [Google Scholar]
- 86.Gronwald J, Byrski T, Huzarski T, et al. Neoadjuvant therapy with cisplatin in BRCA1-positive breast cancer patients. J Clin Oncol. 2009;27(15s suppl):502. doi: 10.1007/s10549-008-0128-9. Abstract. [DOI] [PubMed] [Google Scholar]
- 87.Silver DP, Richardson AL, Eklund AC, et al. Efficacy of neoadjuvant cisplatin in triple-negative breast cancer. J Clin Oncol. 2010;28(7):1145–1153. doi: 10.1200/JCO.2009.22.4725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Bolderson E, Richard DJ, Zhou BB, et al. Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair. Clin Cancer Res. 2009;15(20):6314–6320. doi: 10.1158/1078-0432.CCR-09-0096. [DOI] [PubMed] [Google Scholar]
- 89.McCabe N, Turner NC, Lord CJ, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res. 2006;66(16):8109–8115. doi: 10.1158/0008-5472.CAN-06-0140. [DOI] [PubMed] [Google Scholar]
- 90.Tutt AN, Lord CJ, McCabe N, et al. Exploiting the DNA repair defect in BRCA mutant cells in the design of new therapeutic strategies for cancer. Cold Spring Harb Symp Quant Biol. 2005;70:139–148. doi: 10.1101/sqb.2005.70.012. [DOI] [PubMed] [Google Scholar]
- 91.Plummer ER. Inhibition of poly(ADP-ribose) polymerase in cancer. Curr Opin Pharmacol. 2006;6(4):364–368. doi: 10.1016/j.coph.2006.02.004. [DOI] [PubMed] [Google Scholar]
- 92.James CR, Quinn JE, Mullan PB, et al. BRCA1, a potential predictive biomarker in the treatment of breast cancer. Oncologist. 2007;12(2):142–150. doi: 10.1634/theoncologist.12-2-142. [DOI] [PubMed] [Google Scholar]
- 93.Tuma RS. Combining carefully selected drug, patient genetics may lead to total tumor death. J Natl Cancer Inst. 2007;99(20):1505–1506. 1509. doi: 10.1093/jnci/djm194. [DOI] [PubMed] [Google Scholar]
- 94.Jones C, Plummer ER. PARP inhibitors and cancer therapy: early results and potential applications. Br J Radiol. 2008;81(Spec No 1):S2–5. doi: 10.1259/bjr/30872348. [DOI] [PubMed] [Google Scholar]
- 95.Ashworth A. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. J Clin Oncol. 2008;26(22):3785–3790. doi: 10.1200/JCO.2008.16.0812. [DOI] [PubMed] [Google Scholar]
- 96.Tutt A, Robson M, Garber JE, et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376(9737):235–244. doi: 10.1016/S0140-6736(10)60892-6. [DOI] [PubMed] [Google Scholar]
- 97.Metcalfe KA, Lynch HT, Ghadirian P, et al. The risk of ovarian cancer after breast cancer in BRCA1 and BRCA2 carriers. Gynecol Oncol. 2005;96(1):222–226. doi: 10.1016/j.ygyno.2004.09.039. [DOI] [PubMed] [Google Scholar]
- 98.Frost MH, Schaid DJ, Sellers TA, et al. Long-term satisfaction and psychological and social function following bilateral prophylactic mastectomy. JAMA. 2000;284(3):319–324. doi: 10.1001/jama.284.3.319. [DOI] [PubMed] [Google Scholar]
- 99.Metcalfe KA, Semple JL, Narod SA. Satisfaction with breast reconstruction in women with bilateral prophylactic mastectomy: a descriptive study. Plast Reconstr Surg. 2004;114(2):360–366. doi: 10.1097/01.prs.0000131877.52740.0e. [DOI] [PubMed] [Google Scholar]
- 100.Stefanek ME, Helzlsouer KJ, Wilcox PM, et al. Predictors of and satisfaction with bilateral prophylactic mastectomy. Prev Med. 1995;24(4):412–419. doi: 10.1006/pmed.1995.1066. [DOI] [PubMed] [Google Scholar]
- 101.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. 2005;23(31):7849–7856. doi: 10.1200/JCO.2005.09.233. [DOI] [PubMed] [Google Scholar]
- 102.Geiger AM, West CN, Nekhlyudov L, et al. Contentment with quality of life among breast cancer survivors with and without contralateral prophylactic mastectomy. J Clin Oncol. 2006;24(9):1350–1356. doi: 10.1200/JCO.2005.01.9901. [DOI] [PubMed] [Google Scholar]
- 103.Bresser PJ, Seynaeve C, Van Gool AR, et al. Satisfaction with prophylactic mastectomy and breast reconstruction in genetically predisposed women. Plast Reconstr Surg. 2006;117(6):1675–1682. doi: 10.1097/01.prs.0000217383.99038.f5. discussion 1683-1674. [DOI] [PubMed] [Google Scholar]
- 104.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. 2005;23(28):6890–6898. doi: 10.1200/JCO.2005.02.626. [DOI] [PubMed] [Google Scholar]
- 105.Chen CL, Weiss NS, Newcomb P, et al. Hormone replacement therapy in relation to breast cancer. JAMA. 2002;287(6):734–741. doi: 10.1001/jama.287.6.734. [DOI] [PubMed] [Google Scholar]
- 106.Rossouw JE, Anderson GL, Prentice RL, et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–333. doi: 10.1001/jama.288.3.321. [DOI] [PubMed] [Google Scholar]
- 107.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. 2003;289(24):3243–3253. doi: 10.1001/jama.289.24.3243. [DOI] [PubMed] [Google Scholar]
- 108.Eisen A, Lubinski J, Gronwald J, et al. Hormone therapy and the risk of breast cancer in BRCA1 mutation carriers. J Natl Cancer Inst. 2008;100(19):1361–1367. doi: 10.1093/jnci/djn313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Rebbeck TR, Friebel T, Wagner T, et al. Effect of short-term hormone replacement therapy on breast cancer risk reduction after bilateral prophylactic oophorectomy in BRCA1 and BRCA2 mutation carriers: the PROSE Study Group. J Clin Oncol. 2005;23(31):7804–7810. doi: 10.1200/JCO.2004.00.8151. [DOI] [PubMed] [Google Scholar]
- 110.Anderson GL, Chlebowski RT, Aragaki AK, et al. Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women’s Health Initiative randomised placebo-controlled trial. Lancet Oncol. 2012;13(5):476–486. doi: 10.1016/S1470-2045(12)70075-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Madalinska JB, Essink-Bot ML, de Koning HJ, et al. Health-related quality of life in patients with screen-detected versus clinically diagnosed prostate cancer preceding primary treatment. Prostate. 2001;46(2):87–97. doi: 10.1002/1097-0045(20010201)46:2<87::aid-pros1012>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
- 112.Partridge AH, Gelber S, Peppercorn J, et al. Web-based survey of fertility issues in young women with breast cancer. J Clin Oncol. 2004;22(20):4174–4183. doi: 10.1200/JCO.2004.01.159. [DOI] [PubMed] [Google Scholar]
- 113.Lee S, Schover L, Partridge A, et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol. 2006;24:2917–2931. doi: 10.1200/JCO.2006.06.5888. [DOI] [PubMed] [Google Scholar]
- 114.The Ethics Committee of the American Society for Reproductive. Medicine fertility preservation and reproduction in cancer patients. Fertil Steril. 2005;83(6):1622–1628. doi: 10.1016/j.fertnstert.2005.03.013. [DOI] [PubMed] [Google Scholar]
- 115.Schover LR, Rybicki LA, Martin BA, et al. Having children after cancer. Cancer. 1999;86(4):697–709. doi: 10.1002/(sici)1097-0142(19990815)86:4<697::aid-cncr20>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
- 116.Partridge AH, Gelber S, Peppercorn J, et al. Web-based survey of fertility issues in young women with breast cancer. J Clin Oncol 2004. 2004;22(20):4174–4183. doi: 10.1200/JCO.2004.01.159. [DOI] [PubMed] [Google Scholar]
- 117.Quinn GP, Vadaparampil ST, Lee JH, et al. Physician referral for fertility preservation in oncology patients: a national study of practice behaviors. J Clin Oncol. 2009;27(35):5952–5957. doi: 10.1200/JCO.2009.23.0250. [DOI] [PubMed] [Google Scholar]
- 118.Staton AD, Kurian AW, Cobb K, et al. Cancer risk reduction and reproductive concerns in female BRCA1/2 mutation carriers. Fam Cancer. 2008;7(2):179–186. doi: 10.1007/s10689-007-9171-7. [DOI] [PubMed] [Google Scholar]
- 119.Friedman LC, Kramer RM. Reproductive issues for women with BRCA mutations. J Natl Cancer Inst Monogr. 2005;(34):83–86. doi: 10.1093/jncimonographs/lgi012. [DOI] [PubMed] [Google Scholar]
- 120.Smith KR, Ellington L, Chan AY, et al. Fertility intentions following testing for a BRCA1 gene mutation. Cancer Epidemiol Biomarkers Prev. 2004;13(5):733–740. [PubMed] [Google Scholar]
- 121.Quinn G, Vadaparampil S, Wilson C, et al. Attitudes of high-risk women toward preimplantation genetic diagnosis. Fertil Steril. 2009;91(6):2361–2368. doi: 10.1016/j.fertnstert.2008.03.019. [DOI] [PubMed] [Google Scholar]
- 122.Ogilvie CM, Braude PR, Scriven PN. Preimplantation genetic diagnosis: an overview. J Histochem Cytochem. 2005;53(3):255–260. doi: 10.1369/jhc.4B6395.2005. [DOI] [PubMed] [Google Scholar]
- 123.Baruch S, Kaufman D, Hudson KL. Genetic testing of embryos: practices and perspectives of US in vitro fertilization clinics. Fertil Steril. 2008;89(5):1053–1058. doi: 10.1016/j.fertnstert.2007.05.048. [DOI] [PubMed] [Google Scholar]
- 124.Dayal MB, Athanasiadis I. [Accessed June 28, 2012];Preimplantation Genetic Diagnosis. http://www.emedicine.com/med/topic3520.htm#section~bibliography.
- 125.Offit K, Kohut K, Clagett B, et al. Cancer genetic testing and assisted reproduction. J Clin Oncol. 2006;24(29):4775–4782. doi: 10.1200/JCO.2006.06.6100. [DOI] [PubMed] [Google Scholar]
- 126.Kim SS, Klemp J, Fabian C. Breast cancer and fertility preservation. Fertil Steril. 2011;95(5):1535–1543. doi: 10.1016/j.fertnstert.2011.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Jacobsen PB, Valdimarsdottier HB, Brown KL, et al. Decision-making about genetic testing among women at familial risk for breast cancer. Psychosom Med. 1997;59(5):459–466. doi: 10.1097/00006842-199709000-00001. [DOI] [PubMed] [Google Scholar]
- 128.Sussner KM, Jandorf L, Thompson HS, et al. Interest and beliefs about BRCA genetic counseling among at-risk Latinas in New York City. J Genet Couns. 2010;19(3):255–268. doi: 10.1007/s10897-010-9282-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vadaparampil ST, Quinn GP, Dutil J, et al. A pilot study of knowledge and interest of genetic counseling and testing for hereditary breast and ovarian cancer syndrome among Puerto Rican women. J Community Genet. 2011;2(4):211–221. doi: 10.1007/s12687-011-0058-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.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. 2010;14(1):99–106. doi: 10.1089/gtmb.2009.0088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Offit K, Groeger E, Turner S, et al. The “duty to warn” a patient’s family members about hereditary disease risks. JAMA. 2004;292(12):1469–1473. doi: 10.1001/jama.292.12.1469. [DOI] [PubMed] [Google Scholar]
- 132.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. 2008;12(1):81–91. doi: 10.1089/gte.2007.0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.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) 2004;13(2):169–179. doi: 10.1111/j.1365-2354.2004.00460.x. [DOI] [PubMed] [Google Scholar]
- 134.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. 2007;9(5):275–282. doi: 10.1097/gim.0b013e31804ec075. [DOI] [PubMed] [Google Scholar]
- 135.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. 2006;24(4):700–706. doi: 10.1200/JCO.2005.01.7541. [DOI] [PubMed] [Google Scholar]
- 136.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 C Semin Med Genet. 2003;119C(1):11–18. doi: 10.1002/ajmg.c.10003. [DOI] [PubMed] [Google Scholar]
- 137.Dancyger C, Wiseman M, Jacobs C, et al. Communicating BRCA1/2 genetic test results within the family: a qualitative analysis. Psychol Health. 2011;26(8):1018–1035. doi: 10.1080/08870446.2010.525640. [DOI] [PubMed] [Google Scholar]
- 138.Loud JT, Weissman NE, Peters JA, et al. Deliberate deceit of family members: a challenge to providers of clinical genetics services. J Clin Oncol. 2006;24(10):1643–1646. doi: 10.1200/JCO.2005.02.6203. [DOI] [PubMed] [Google Scholar]
- 139.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. 2007;25(24):3705–3711. doi: 10.1200/JCO.2006.09.1900. [DOI] [PubMed] [Google Scholar]
- 140.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. 2009;18(2):200–208. doi: 10.1002/pon.1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Sharff ME, Demarco TA, Mays D, et al. Parenting through genetic uncertainty: themes in the disclosure of breast cancer risk information to children. Genet Test Mol Biomarkers. 2012;16(5):376–382. doi: 10.1089/gtmb.2011.0154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.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. 1995;57(5):1233–1241. [PMC free article] [PubMed] [Google Scholar]
- 143.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. 2010;28(5):893–901. doi: 10.1200/JCO.2009.27.0660. [DOI] [PubMed] [Google Scholar]
- 144.Committee on Bioethics. Ethical issues with genetic testing in pediatrics. Pediatrics. 2001;107(6):1451–1455. doi: 10.1542/peds.107.6.1451. [DOI] [PubMed] [Google Scholar]
- 145.Wertz DC, Fanos JH, Reilly PR. Genetic testing for children and adolescents. Who decides? JAMA. 1994;272(11):875–881. [PubMed] [Google Scholar]
- 146.Leib JR, Hoodfar E, Haidle JL, et al. The new genetic privacy law. Commun Oncol. 2008;5(6):124–127. [Google Scholar]
- 147.Baruch S, Hudson K. Civilian and military genetics: nondiscrimination policy in a post-GINA world. Am J Hum Genet. 2008;83(4):435–444. doi: 10.1016/j.ajhg.2008.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.American Cancer Society. [Accessed June 28, 2012];Cancer Facts & Figures. 2011 http://www.cancer.org/Research/CancerFactsFigures/index.
- 149.Vos J, Jansen AM, Menko F, et al. Family communication matters: the impact of telling relatives about unclassified variants and uninformative DNA-test results. Genet Med. 2011;13(4):333–341. doi: 10.1097/GIM.0b013e318204cfed. [DOI] [PubMed] [Google Scholar]
- 150.Vos J, Menko F, Jansen AM, van Asperen CJ, et al. A whisper-game perspective on the family communication of DNA-test results: a retrospective study on the communication process of BRCA1/2-test results between proband and relatives. Fam Cancer. 2011;10(1):87–96. doi: 10.1007/s10689-010-9385-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Vos J, Oosterwijk JC, Gomez-Garcia E, et al. Perceiving cancer-risks and heredity-likelihood in genetic-counseling: how counselees recall and interpret BRCA 1/2-test results. Clin Genet. 2011;79(3):207–218. doi: 10.1111/j.1399-0004.2010.01581.x. [DOI] [PubMed] [Google Scholar]
- 152.Berliner JL, Fay AM. Practice Issues Subcommittee of the National Society of Genetic Counselors’ Familial Cancer Risk Counseling Special Interest Group. Risk assessment and genetic counseling for hereditary breast and ovarian cancer: recommendations of the National Society of Genetic Counselors. J Genet Couns. 2007;16(3):241–260. doi: 10.1007/s10897-007-9090-7. [DOI] [PubMed] [Google Scholar]
- 153.National Cancer Policy Forum; Institute of Medicine. National Academy of Sciences; Washington, DC: 2007. [Accessed June 28, 2012]. Cancer-Related Genetic Testing and Counseling: Workshop Proceedings. http://www.nap.edu/catalog/11971.html. [Google Scholar]
- 154.Riley BD, Culver JO, Skrzynia C, et al. Essential elements of genetic cancer risk assessment, counseling, and testing: updated recommendations of the National Society of Genetic Counselors. J Genet Couns. 2012;21(2):151–161. doi: 10.1007/s10897-011-9462-x. [DOI] [PubMed] [Google Scholar]
- 155.American College of Obstetricians and Gynecologists; ACOG Committee on Practice Bulletins: Gynecology; ACOG Committee on Genetics; Society of Gynecologic Oncologists. ACOG Practice Bulletin No. 103: Hereditary breast and ovarian cancer syndrome. Obstet Gynecol. 2009;113(4):957–966. doi: 10.1097/AOG.0b013e3181a106d4. [DOI] [PubMed] [Google Scholar]
- 156.Oncology Nursing Society. [Accessed June 28, 2012];The Role of the Oncology Nurse in Cancer Genetic Counseling. 2009 http://www.ons.org/Publications/Positions/Genetic-Counseling/
- 157.Radford C. Understanding the Evolution of and Term “Genetic Counselor”: Is the Term Being Used Appropriately in Your Community? Oncol Nurse. 2011;4(5):24–25. http://www.theoncologynurse.com/article/understanding-evolution-and-term-%E2%80%9Cgenetic-counselor%E2%80%9D-term-being-used-appropriately-your-comm. [Google Scholar]
- 158.Geier LJ, Mulvey TM, Weitzel JN. Clinical cancer genetics remains a specialized area: how do I get there from here? 2009 Educational Book. American Society of Clinical Oncology; [Accessed June 29, 2012]. http://www.asco.org/ASCOv2/Home/Education%20&%20Training/Educational%20Book/PDF%20Files/2009/09EdBk.PractMgmt.03.pdf. [Google Scholar]
- 159.Cohen SA, McIlvried D, Schnieders J. A collaborative approach to genetic testing: a community hospital’s experience. J Genet Couns. 2009;18(6):530–533. doi: 10.1007/s10897-009-9243-y. [DOI] [PubMed] [Google Scholar]
- 160.DeMarco TA, Smith KL, Nusbaum RH, et al. Practical aspects of delivering hereditary cancer risk counseling. Semin Oncol. 2007;34(5):369–378. doi: 10.1053/j.seminoncol.2007.07.003. [DOI] [PubMed] [Google Scholar]
- 161.MacDonald DJ, Blazer KR, Weitzel JN. Extending comprehensive cancer center expertise in clinical cancer genetics and genomics to diverse communities: the power of partnership. J Natl Compr Canc Netw. 2010;8(5):615–624. doi: 10.6004/jnccn.2010.0046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Esserman L, Kaklamani V. Lessons learned from genetic testing. JAMA. 2010;304(9):1011–1012. doi: 10.1001/jama.2010.1263. [DOI] [PubMed] [Google Scholar]
- 163.Brierley KL, Campfield D, Ducaine W, et al. Errors in delivery of cancer genetics services: implications for practice. Conn Med. 2010;74(7):413–423. [PubMed] [Google Scholar]
- 164.Lawrence WF, Peshkin BN, Liang W, et al. Cost of genetic counseling and testing for BRCA1 and BRCA2 breast cancer susceptibility mutations. Cancer Epidemiol Biomarkers Prev. 2001;10(5):475–481. [PubMed] [Google Scholar]
- 165.Schwartz GF, Hughes KS, Lynch HT, et al. Proceedings of the international consensus conference on breast cancer risk, genetics, & risk management, April, 2007. Cancer. 2008;113(10):2627–2637. doi: 10.1002/cncr.23903. [DOI] [PubMed] [Google Scholar]
- 166.Hampel H, Sweet K, Westman JA, et al. Referral for cancer genetics consultation: a review and compilation of risk assessment criteria. J Med Genet. 2004;41(2):81–91. doi: 10.1136/jmg.2003.010918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Weitzel JN. Genetic cancer risk assessment. Putting it all together. Cancer. 1999;86(11 suppl):2483–2492. doi: 10.1002/(sici)1097-0142(19991201)86:11+<2483::aid-cncr5>3.3.co;2-w. [DOI] [PubMed] [Google Scholar]
- 168.Ganz PA, Kwan L, Somerfield MR, et al. The role of prevention in oncology practice: results from a 2004 survey of American Society of Clinical Oncology members. J Clin Oncol. 2006;24(18):2948–2957. doi: 10.1200/JCO.2006.05.8321. [DOI] [PubMed] [Google Scholar]
- 169.McPherson E, Zaleski C, Benishek K, et al. Clinical genetics provider real-time workflow study. Genet Med. 2008;10(9):699–706. doi: 10.1097/gim.0b013e318182206f. [DOI] [PubMed] [Google Scholar]
- 170.Bernhardt BA, Tumpson JE, Pyeritz RE. The economics of clinical genetics services. IV. Financial impact of outpatient genetic services on an academic institution. Am J Hum Genet. 1992;50(1):84–91. [PMC free article] [PubMed] [Google Scholar]
- 171.Vadaparampil ST, Quinn GP, Miree CA, et al. Recall of and reactions to a surgeon referral letter for BRCA genetic counseling among high-risk breast cancer patients. Ann Surg Oncol. 2009;16(7):1973–1981. doi: 10.1245/s10434-009-0479-4. [DOI] [PubMed] [Google Scholar]
- 172.Vadaparampil ST, Quinn GP, Brzosowicz J, et al. Experiences of genetic counseling for BRCA1/2 among recently diagnosed breast cancer patients: a qualitative inquiry. J Psychosoc Oncol. 2008;26(4):33–52. doi: 10.1080/07347330802359586. [DOI] [PubMed] [Google Scholar]
- 173.Vadaparampil ST, Quinn GP, Lee JH, et al. Satisfaction with physician recommendation for and information about genetic counseling among breast cancer patients. Breast J. 2011;17(1):79–82. doi: 10.1111/j.1524-4741.2010.01027.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Acheson LS, Stange KC, Zyzanski S. Clinical genetics issues encountered by family physicians. Genet Med. 2005;7(7):501–508. doi: 10.1097/01.gim.0000177418.24176.9b. [DOI] [PubMed] [Google Scholar]
- 175.Vig HS, Armstrong J, Egleston BL, et al. Cancer genetic risk assessment and referral patterns in primary care. Genet Test Mol Biomarkers. 2009;13(6):735–741. doi: 10.1089/gtmb.2009.0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.(ASCO) ASoCO. Cancer Genetics and Cancer Predisposition Testing. 2. Alexandria, VA: American Society of Clinical Oncology Publishing; 2004. [Google Scholar]
- 177.National Cancer Policy Forum, Institute of Medicine. Cancer-Related Genetic Testing and Counseling: Workshop Proceedings. The National Academies Press; 2007. [Google Scholar]
- 178.Daly MB, Axilbund JE, Buys S, et al. Genetic/familial high-risk assessment: breast and ovarian. J Natl Compr Canc Netw. 2010;8(5):562–594. doi: 10.6004/jnccn.2010.0043. [DOI] [PubMed] [Google Scholar]
- 179.Calzone KA, Jenkins J, Masny A. Core competencies in cancer genetics for advanced practice oncology nurses. Oncol Nurs Forum. 2002;29(9):1327–1333. doi: 10.1188/02.ONF.1327-1333. [DOI] [PubMed] [Google Scholar]
- 180.Genetic testing in adoption. The American Society of Human Genetics Social Issues Committee and The American College of Medical Genetics Social, Ethical, and Legal Issues Committee. Am J Hum Genet. 2000;66(3):761–767. [PMC free article] [PubMed] [Google Scholar]
- 181.Kolb SE, Aguilar MC, Dinenberg M, Kaye CI. Genetics education for primary care providers in community health settings. J Community Health. 1999;24(1):45–59. doi: 10.1023/a:1018765215470. [DOI] [PubMed] [Google Scholar]
- 182.Korf BR. Medical education in the ‘postgenomic era’. Postgrad Med. 2000;108(3):15–18. doi: 10.3810/pgm.2000.09.1.1219. [DOI] [PubMed] [Google Scholar]
- 183.Peterson SK, Rieger PT, Marani SK, et al. Oncology nurses’ knowledge, practice, and educational needs regarding cancer genetics. Am J Med Genet. 2001;98(1):3–12. [PubMed] [Google Scholar]
- 184.Reynolds PP, Benkendorf JL. Genes and generalists: why we need professionals with added competencies. West J Med. 1999;171(5–6):375–379. [PMC free article] [PubMed] [Google Scholar]
- 185.Schwartz MD, Valdimarsdottir HB, DeMarco TA, et al. Randomized trial of a decision aid for BRCA1/BRCA2 mutation carriers: impact on measures of decision making and satisfaction. Health Psychol. 2009;28(1):11–19. doi: 10.1037/a0013147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Ozanne EM, Loberg A, Hughes S, et al. Identification and management of women at high risk for hereditary breast/ovarian cancer syndrome. Breast J. 2009;15(2):155–162. doi: 10.1111/j.1524-4741.2009.00690.x. [DOI] [PubMed] [Google Scholar]
- 187.Drohan B, Ozanne EM, Hughes KS. Electronic health records and the management of women at high risk of hereditary breast and ovarian cancer. Breast J. 2009;15 (suppl 1):S46–S55. doi: 10.1111/j.1524-4741.2009.00796.x. [DOI] [PubMed] [Google Scholar]
- 188.Drohan B, Roche CA, Cusack JC, Jr, et al. Hereditary breast and ovarian cancer and other hereditary syndromes: using technology to identify carriers. Ann Surg Oncol. 2012;19(6):1732–1737. doi: 10.1245/s10434-012-2257-y. [DOI] [PubMed] [Google Scholar]
- 189.Zon RT, Goss E, Vogel VG, et al. American Society of Clinical Oncology policy statement: the role of the oncologist in cancer prevention and risk assessment. J Clin Oncol. 2009;27(6):986–993. doi: 10.1200/JCO.2008.16.3691. [DOI] [PMC free article] [PubMed] [Google Scholar]
