Open access peer-reviewed chapter

Breast Cancer and BRCA1 and BRCA2 Pathogenic Variants

Written By

Naren Basu and D. Gareth Evans

Submitted: 14 June 2019 Reviewed: 20 December 2019 Published: 20 January 2020

DOI: 10.5772/intechopen.90926

From the Edited Volume

Breast Cancer and Breast Reconstruction

Edited by Luis Tejedor, Susana Gómez Modet, Lachezar Manchev and Arli Aditya Parikesit

Chapter metrics overview

896 Chapter Downloads

View Full Metrics

Abstract

Breast cancer remains the most common female cancer worldwide. The majority will arise spontaneously, with almost a third having a heritable component. Approximately 5–10% of all breast cancers will have a strong inherited element with pathogenic variants in the BRCA1 and BRCA2 amongst the most studied breast cancer genes. An overview of breast cancer is provided with references to the clinical and pathological features in BRCA1 and BRCA2 related cancers. The roles of PARP inhibitors and immunotherapy are discussed. The management of healthy individuals harbouring a pathogenic variant in the two genes is reviewed and future directions considered.

Keywords

  • BRCA1
  • BRCA2
  • breast cancer
  • risk reduction
  • mastectomy
  • pathogenic variant

1. Introduction

Breast cancer remains the most common cancer amongst women in the world [1]. In the UK, 25% of all female cancers originate from the breast with estimates that 1 in 7–10 will develop this disease during their lifetime [2]. In 2018, there were over 2 million new cases of breast cancer worldwide, accounting for almost 12% of all cancer [3]. In developed countries, almost 80% of breast cancers occur in post-menopausal women, predominantly in the Caucasian population.

Breast cancer represents a heterogenous disease process and an understanding of the clinical aspects is required prior to appreciating the aspects of breast cancer amongst BRCA1/2 pathogenic variant (PV) carriers.

Advertisement

2. Invasive or in-situ (non-invasive cancer)

The vast majority of breast cancers are invasive (up to 90%) presenting via a symptomatic pathway. In contrast, in-situ disease tends to be more commonly identified via breast screening programmes (e.g., NHSBSP) with advances in digital mammography or incidentally. In the USA, in situ disease comprises almost 25% of all breast cancers, 80% of which are identified by breast screening [4].

In histopathological terms, invasive cancers will have breached the basement membrane with metastatic potential compared with the more innocuous in-situ disease where the disease process is contained within the basement membrane, with theoretically limited or no metastatic potential.

Advertisement

3. Ductal or lobular origin

The microanatomy of the breast can be considered to comprise of terminal ductal lobular unit [5, 6]. Each breast lobule is by a collecting duct terminating in the lobule. This serves as the basic functional unit of the breast. Invasive ductal carcinoma (also termed no special type carcinoma) represents the most common type of breast cancer accounting for almost 80% of cancer. Invasive lobular carcinoma is less common accounting for almost 10% of cases.

These two types of breast cancer are biologically different. Invasive lobular cancers tend to have a more spreading growth pattern, meaning that they can be more difficult to diagnose or size on routine mammograms or clinical examination. In addition, they may be resistant to neo-adjuvant chemotherapy. Invasive lobular cancers tend to be more hormone-sensitive, HER2 negative and lower grade breast cancer (see below).

Advertisement

4. Grade of breast cancer

This refers to the microscope assessment of the breast cancer and determining how the cells look compared with normal breast tissue [6]. This in turn will predict the biological activity of the breast cancer, with high-grade tumours considered to be faster growing with a more aggressive behaviour.

4.1 Grades in in-situ disease

  1. Low grade—slow growing and more closely resemble normal breast tissue.

  2. Moderate/intermediate grade—more abnormal looking compared with low grade, with a more rapid growth pattern.

  3. High grade—cells look quite different to normal breast tissue, grow quicker with an increased chance of progressing to an invasive cancer.

4.2 Grades in invasive cancer

There are three different grades of invasive breast cancer based on an assessment of tubule formation, nuclear pleomorphism and mitotic count:

  1. Grade 1 (well differentiated).

  2. Grade 2 (moderately differentiated).

  3. Grade 3 (poorly differentiated).

Advertisement

5. Hormone receptor status

Oestrogen receptors (ER) were first identified in 1958 (Elwood, Chicago) but an appreciation of the hormonal component of breast cancer predates this to the 1890s when George Beatson performed oophorectomy as a treatment for breast cancer in women [7]. It still remains a critical biological factor in the modern day management of breast cancer.

The oestrogen (ER) and progesterone receptors (PR) relevant to breast cancer are intracellular receptors that are activated by the hormone oestrogen (17 beta oestrodiol) acting as a DNA-binding transcription factor, in particular stimulation of mammary cells.

Approximately 70–80% of breast cancers overexpress ER [8]. These hormone sensitive breast cancers may be treated with anti-hormone therapy by either selective oestrogen receptor modulators (e.g., Tamoxifen) or aromatase inhibitors.

Advertisement

6. HER2 receptor status

The HER2 receptor (human epidermal growth factor receptor) is a transmembrane tyrosine kinase that is overexpressed in 15–20% of breast cancers [9]. Amplification of this oncogene is considered a marker of aggressiveness and is an important biomarker for targeted therapy. Drugs targeting the HER2 receptor (e.g., Trastuzumab, Pertuzumab) have significantly altered the clinical outcomes of otherwise poor prognosis HER2 positive breast cancers.

Advertisement

7. Molecular subtypes of breast cancer

Gene expression profiling has enabled molecular classification of breast cancer into intrinsic subtypes [10] that include:

  1. Luminal A: hormone receptor positive (ER/PR +ve) and HER2 negative. They express low levels of the protein Ki-67, a cellular marker for proliferation. These cancers tend to be low grade, slow growing, good prognosis cancers.

  2. Luminal B: hormone receptor positive (ER/PR +ve) and either HER2 positive or negative with a high level of Ki-67 expression. Faster growing and worse prognosis than luminal A cancers.

  3. Triple negative breast cancer (TNBC)/basal-like: these are hormone receptor negative and HER2 negative (ER/PR/HER2 −ve). This accounts for 10–20% of all breast cancers, more commonly found in women with a BRCA1 PV, young women and African-American women. It is a rapidly growing, poor prognosis cancer.

  4. HER2-enriched: these are HER2 positive and ER/PR negative cancer (HER2 +ve, ER/PR −ve). They tend to grow faster than luminal cancers but can be treated with targeted therapies.

Advertisement

8. Treatments for breast cancer

The management of breast cancer is a multi-modal delivered in a multi-disciplinary setting. In brief, treatments include:

8.1 Surgery

Excision of the breast lesion and assessment of the axillary lymph node for regional metastasis. Historically this involved mastectomy with axillary clearance, which has now been refined following some seminal trial in breast cancer surgery to offering appropriate women breast conserving surgery (“lumpectomy”) and sentinel node biopsy. There are still important indications for mastectomy that would include genetic mutation status, family history, previously treated breast cancer, inflammatory breast cancer and patient choice.

8.2 Anti endocrine therapy

Hormone sensitive breast cancers are treated with anti hormonal therapy for a minimum period of 5 years that can be extended to 10–15 years based on recent emerging data. There is an improved overall survival, reduced recurrence rate and reduction in the incidence of a contralateral breast cancer.

8.3 Radiotherapy

This is a localised, targeted therapy of high-energy X-rays that complements breast conservation surgery in almost all cases of invasive cancer, reducing local recurrence rates by almost 20–30% [11]. It may be offered following mastectomy if there is a high burden of disease.

8.4 Chemotherapy

This systemic treatment may be offered pre surgery (neo adjuvant) or post surgery (adjuvant). The choice of chemotherapy regimen is determined by number factors and may include anthracycline-taxane combinations. Perceived benefits include an improvement in survival, reduction in local recurrence and downstaging of tumour burden.

8.5 Anti HER2 treatment

HER2 targeted immunotherapy consists of monoclonal antibodies (e.g., Trastuzumab)—previously discussed.

8.6 Other treatments

PARP inhibitors: poly ADP Ribose Polymerase inhibitors are targeted therapies with increasing use in patients with a PV in the BRCA1/2 gene. This is discussed in more detail later.

Immunotherapy: this involves stimulating the host immune system actively immunisation (use of cancer vaccine) or passive immunisation (use of tumour specific antibodies or immune modulators). Of particular growing importance is the identification of patients with TNBC overexpressing immune evasion molecules (e.g., PDL: programmed death ligand) that may offer an additional treatment modality in this poor prognosis group [12].

Advertisement

9. Breast cancer genetics

The majority of breast cancers arise spontaneously—with known risk factors that include oestrogen exposure, reproductive history (late first or no pregnancy) and lifestyle practices. However, a significant proportion (20–30%) of cancers will have an inherited component, with family history remaining the strongest risk factor [13]. These tend to be classed as germline PVs in contrast to somatic mutations—arising from the interactions between environment and genetics.

Approximately 5–10% of breast cancer will have a strong inherited component that can be further sub-divided [13] according to strength of penetrance:

9.1 High penetrance genes

  • BRCA1

  • BRCA2

  • PALB2

  • TP53

  • PTEN

  • STK11

  • CDH1

9.2 Moderate penetrance genes

  • ATM

  • CHEK2

  • NF1

  • BARD1

9.3 Low penetrance genes

The advent of Genome wide association studies has rapidly enabled the identification of over 100 breast cancer single nucleotide polymorphisms [14] that confer a small individual increase or decrease in breast cancer risk, but appear to work in a polygenic, multiplicative pattern contributing to a more significant risk.

Advertisement

10. Breast cancer and the BRCA1 gene

This tumour suppressor gene was first identified in 1990 and cloned 4 years later in the US [15]. During the next two decades a bitter commercial battle ensued regarding patenting of the genomic DNA sequence to both BRCA1 and BRCA2. Ultimately, the 2013 US Supreme Court (Association of Molecular Pathology vs. Myriad Genetics) ruled that “isolation of genes found in nature do not render them patentable” [16].

The human BRCA1 gene (Chromosome 17—codes for the breast cancer type 1 susceptibility protein) is involved in non error prone homologous DNA repair. The majority of PVs are frameshift resulting in a truncated protein with founder PVs in the Ashkenazi Jewish populations and Eastern European population [13].

10.1 Penetrance and lifetime risk

The inheritance pattern is autosomal dominant with birth incidence of a BRCA1 PV estimated at 1 in 500–900, accounting for 7–10% of familial breast cancer [13].

Harbouring a BRCA1 PV confers a lifetime risk of breast cancer of approximately 60–85% [17, 18] and up to 60% for developing ovarian cancer (usually high-grade serous carcinomas). Risk of breast cancer is inversely proportional to age—a trend not so clearly seen in ovarian cancer (i.e., not age dependant). A recent prospective cohort study of 6036 BRCA1 PV carriers [19] identified that the breast cancer incidences per decade of age increased from 21–30 years to 31–40 years but then remained at 23.5–28.3 per 1000 person years from age 31 to 70 years. Therefore, there is a rapid increase in breast cancer incidence in early childhood that plateaus and remains constant throughout the remaining adult life.

Family history of breast cancers amongst first and second-degree relatives increased the relative risk breast cancer, a trend not seen in ovarian cancer risk in families with ovarian cancer. BRCA1 PVs located outside the region bounded by c.2282 to c.4071 were associated with the highest risk of developing breast cancer [19].

There is a significant risk of developing a contralateral breast cancer once diagnosed with breast cancer. Our own studies have confirmed that this risk is approximately 2–3% per year, persists for at least 30 years.

Other cancers associated with this PV include pancreatic cancer (RR 2.26), uterine body and cervical cancer (RR 2.65, RR 3.72, respectively) and prostate cancer in the under 65s (1.82) [13].

10.2 Pathology of BRCA1 cancers

BRCA1 related breast cancers are heterogenous but have some important clinical features. They are most often triple negative approximately 70–80% (ER, PR, Her2 −ve) with expression of basal markers (CK5/6, CK14, SMA, P Cadherin, EGFR0). Histologically, they are similar to high-grade medullary carcinomas with pushing margins, high mitotic counts and lymphocytic infiltrate [20].

The international collaborative study (CIMBA) assessed pathological characteristics amongst 3797 BRCA1 carriers [21] and identified that the mean age of breast cancer was 40 years. The majority of breast cancers were of the ductal, no-special type variety with the majority of cancers demonstrating a basal-triple negative phenotype (78% of tumours were ER-negative, 79% were PR-negative, 90% HER2-negative and 68% TNBC). High-grade tumours were most common, with a clear relationship between age at diagnosis and grade of breast cancer—grade decreased with increasing age.

11. Breast cancer and the BRCA2 gene

This DNA repair gene was localised in the UK in 1994 and identified (cloned) in 1995 [22]. It is situated on Chromosome 13 at position 12.3 (13q12.3). Although the structures of the BRCA1 and BRCA2 gene vary from each other, there is some functional overlap. BRCA1 associates with BRCA2 through PALB2, a major binding partner of BRCA2. There is some data to suggest that BRCA1 acts as an upstream regulator of BRCA2 as BRCA1 promotes the concentration of PALB2 and BRCA2 at DNA damage sites [23].

11.1 Penetrance and lifetime risk

PVs in BRCA2 are nearly all inherited suggesting a large founder effect. This is important for practical purposes as certain populations can be tested for known PVs (e.g., single PV 999del5 accounts for almost all inherited breast and ovarian cancer in Iceland) [13].

Lifetime breast cancer risk has a wider range (40–85%) compared with BRCA1, and a slightly lower risk of developing ovarian cancer (20–30%) [13]. Approximately 1 in 400–800 women carry a PV (outbred population) that accounts for 10% of familial breast/ovarian cancer [14].

Several other cancers are more commonly associated with these PV: cholangiocarcinoma, melanoma, pancreatic cancer (overall RR 4.1), gastric cancer (RR 2.7) and prostate cancer (10–20% life time risk) [13]. Approximately 10% of male breast cancer is associated with BRCA2, a trend not seen in BRCA1 PVs.

11.2 Pathology of BRCA2 cancers

Breast cancer in BRCA2 PVs carriers is a more heterogeneous group compared with BRCA1 carriers, with more semblance to sporadic breast cancers. Pathology shows that these breast cancers are more often ER+ve compared with controls, with some studies showing increased DCIS and lobular cancer [20].

A study of breast cancer amongst 6893 BRCA1 and BRCA2 PV carriers found similar proportion of ductal, no-special type cancers (approximately 80%) with a four-fold increase in lobular cancers amongst BRCA2 carriers (8% compared with 2% in BRCA1 patients) [21]. Overall, there were double the number of Grade 1 and 2 cancers in the BRCA2 compared with BRCA1, with more Grade 3 cancers in the BRCA1 group (77% compared with 50% in BRCA2 carriers).

There is paucity of data regarding the pre-invasive progression pathway amongst BRCA1/2 carriers. Whereas a DCIS associated pre-malignant pathway has been postulated in sporadic cancers, this has not been shown in BRCA1/2 PV carriers. There is conflicting data regarding incidence of DCIS in this patient group. Some reports suggest an increased incidence of DCIS identified in risk-reducing mastectomy specimens as opposed to other studies identifying DCIS less frequently near the invasive cancer compared with sporadic tumours [24].

12. Prognosis for BRCA mutated breast cancer

The POSH study (Prospective Outcomes in Sporadic versus Hereditary breast cancer), designed as a prospective cohort study, assessed the outcomes of women aged 40 years or below diagnosed with an invasive breast cancer in the UK [25]. With a follow up of nearly 10 years, 12% of all patients were identified with a PV in either the BRCA1/2 gene. Multivariate analysis identified no overall difference in survival between BRCA positive or BRCA negative patients. Amongst patients with TNBC, there was a short-term survival benefit at 2 years amongst BRCA carriers. The authors hypothesised that this short-lived benefit may reflect greater sensitivity of BRCA mutant cancer to chemotherapy and/or greater visibility of BRCA related cancers to the immune system [25].

Studies prior to the POSH studied inconsistent effects of BRCA1/2 PV status on breast cancer outcomes. A meta-analysis of 66 relevant studies [26] did not identify inferior breast cancer outcomes amongst BRCA1/2 PV carriers despite the biological differences in breast cancer type already discussed.

In addition these women have similar survival whether they are treated with breast conservation surgery or a mastectomy, despite having significant higher rates of local failure with breast conservation [27]. An international longitudinal study of 655 patients with BRCA1/2 PVs showed no differences in overall survival, distant or regional recurrence at 20 years whether women underwent a mastectomy or breast conservation therapy with radiotherapy. Interestingly, most local recurrences are second primary breast cancers rather than failure to control the primary breast cancer [27].

12.1 Radiotherapy

The effect of radiotherapy on BRCA1/2 associated breast cancer merits consideration. These cancers are characterised by defects in homologous recombination, resulting in inadequate repair of double stranded DNA breaks—a hallmark of PVs in the BRCA1/2 genes. In particular, younger patients with a higher rate of cell proliferation may be more susceptible to the carcinogenic effects of radiotherapy. Studies have confirmed an increased risk of contralateral breast cancer amongst young patients with a sporadic breast cancer treated with radiotherapy suggesting an effect of low-dose scatter radiation to surround healthy tissue [28].

In patients with a BRCA1/2 PV associated breast cancer treated with unilateral radiotherapy, hypothetically there should be an increased risk of contralateral breast cancers particularly in the younger patient group, as the contralateral healthy breast will receive some scatter radiation. In addition, adjuvant radiotherapy to the index breast of BRCA1/2 PV carriers should be more efficacious compared with sporadic cancer by virtue of the aberration in DNA repair. A recent Dutch study was not able to demonstrate any association between radiotherapy and contralateral breast cancer risk [29]. They observed a growing trend in their population group of BRCA1/2 PV carriers away from breast conservation with radiotherapy towards mastectomy and contralateral mastectomy.

12.2 Chemotherapy

Healthy, normal breast tissue exposed to toxic, chemotherapeutic agents will result in DNA damage that is then partly repaired by the BRCA1/2 DNA damage response. Different chemotherapy agents used to treat breast cancers exhibit differences in mechanism of action. Taxane-based agents work by disrupting the microtubule function whereas anthracyclines induce topoisomerase II mediated toxicity, DNA intercalation and generation of reactive oxidative species. In vitro studies have shown that platinum based chemotoxic agents have a greater sensitivity for BRCA1 mutated cell lines which may in part be due to the disruption of DNA [30].

12.3 PARP inhibitors

This class of targeted therapy work on the principle of “synthetic lethality” [31], whereby a defect in one gene/protein results in cell survival however when synthesised with another gene/protein results in cell death.

Poly ADP Ribose Polymerase (PARP1) is an important protein that repairs single strand DNA breaks. Drugs that inhibit this protein (PARP inhibitors) result in multiple breaks in double stranded DNA that cannot be repaired, resulting in cell death. A proof of concept international study [32] showed a favourable therapeutic index for Olaparib (an orally active PARP inhibitor) amongst patients with advanced or recurrent breast cancer who harboured a BRCA1/2 PV. The OlympiAD study, an international, randomised, Phase III trial recently reported an improved progression free survival amongst patients with metastatic breast cancer who were HER2 negative with a germline PV in either BRCA1/2 [33]. Future directions of research will include combining PARP inhibitors with radiotherapy and platinum based agents [34].

12.4 Risk-reducing surgery

Bilateral risk-reducing mastectomy (BRRM) offers the greatest magnitude of overall risk reduction in health patients harboring a BRCA1/2 PV by removing in excess of 90% of the breast tissue. A recent Cochrane review showed that BRRM reduces the risk of developing breast cancer by 85–100 [35] in high-risk patients. It also showed that BRRM reduced the risk of dying from breast cancer by 81–100% in high-risk patients and 100% in the moderate risk group.

Uptake of BRRM varies amongst women in this high-risk group. From a patient’s perspective, young age and motherhood seem to be positive predictors for choosing surgery [36, 37, 38]. Several studies have shown international variations in uptake of BRRM with the highest rates in the UK and the Netherlands (33–50%) and the US (36%) compared with Poland (3%) and Israel (4%) [39, 40]. Differences in culture, healthcare systems and access to genetic testing are likely to contribute to these differences. However, wide variations are found within countries—for example in three different Canadian regions, the range of uptake was from 8 to 46% [41]—suggesting that other factors are important.

Media coverage and public interest in BRRM has been heightened since 2013 when Angelina Jolie’s revealed her personal experience of bilateral mastectomies based on her inheriting a pathogenic BRCA1 PV from her late mother. The so-called “Angelina Jolie Effect” ensued with increased uptake of genetic testing and BRRM in the US and UK [42, 43, 44].

12.5 Bilateral risk-reducing salpingo oophorectomy (BRRSO)

Women choosing this procedure will reduce their risk of developing ovarian by almost 90%. In addition, there appears to be a reduction in their subsequent risk of developing breast cancer (premenopausal women) [45, 46], with several previous studies showing a risk-reduction of almost 50%. Unlike breast cancer, surveillance for ovarian cancer is limited to measurements of tumour markers (CA125) and transvaginal ultra-sound—both of which may lack sensitivity. As such, uptake of BRRSO is high (up to 75%).

BRRSO renders women post menopausal (surgical menopause) with additional risks to the cardiovascular and skeletal system. In addition, most women are recommended to have completed their family prior to considering this surgery. The climacteric symptoms following this procedure can be debilitating and many women will consider use of HRT to combat these symptoms. Use of HRT itself may increase the risk of developing breast cancer in these women already deemed at high-risk.

The risk reduction for breast cancer has recently been reassessed for potential selection bias [47]. This study used the same methodology previously described [45] to study a cohort of Dutch BRCA1/2 healthy carriers and found that following BRRSO, the incidence of breast cancer was almost halved (Hazard Ratio 0.36–0.62). A revised analysis taking into account the various biases described above showed no real protective effect of BRRSO on breast cancer development (Hazard Ratio 1.09). This has important clinical implications when considering the variations of uptake of BRRM compared with BRRSO—women only choosing BRRSO over BRRM may have had their breast cancer risk reduction overestimated if they only chose BRRSO.

12.6 Chemoprevention

In the UK, healthy women with a BRCA1/2 PV may consider three medications to reduce their risk:

Tamoxifen: this selective oestrogen receptor modulator (first generation) has been shown to reduce the risk in asymptomatic women by approximately 40–50% [48]. In women with breast cancer, tamoxifen has a similar risk reduction in developing CBC. The side-effect profile (hot flushes, increased incidence of endometrial cancer, thromboembolic phenomenon) is an important consideration as less than 15% of women will choose this and remain compliant [49]. Tamoxifen may be considered in pre and post menopausal women.

Raloxifene (second generation SERM), used in the prevention and treatment of osteoporosis has a much better side effect profile with no increase in endometrial cancers but increases thromboembolic risk similar to tamoxifen. Risk reduction was inferior to tamoxifen over an extended follow [50]. Raloxifene is considered in the post-menopausal setting only.

Aromatase inhibitor: Exemestane has been shown to offer 65% relative risk reduction in post-menopausal women at increased risk with a minimal side-effect profile [51]. The IBIS II trial (International Breast Cancer Intervention Studies) randomised post-menopausal women with an increased risk of breast cancer to anastrozole or placebo. At 5 years follow-up, 2% of women taking Anastrozole had developed breast cancer compared with 4% in the placebo group [52].

Although there is not good quality evidence to show that endocrine therapy is not effective in BRCA1 as 70–80% of breast cancers are triple negative and these medications only reduce ER positive cancers they may have less efficacy in BRCA1 PV carriers.

12.7 Lifestyle

Worldwide, the number of oestrogen receptor positive breast cancers is increasing with a reverse pattern seen with oestrogen negative cancer. In developed countries, hormone sensitive breast cancer is particularly amenable to lifestyle prevention—with recent studies suggesting that modification of lifestyle measure may prevent up to 30% of breast cancers [53].

Amongst BRCA carriers, smoking, increased weight and reduced physical activity further increases the risk of breast cancer. These lifestyle measures need to commence in adolescence and adherence to this (150 min weekly activity, BMI < 25, <1 alcoholic drink daily) has been shown to reduce mortality in BRCA carriers by almost 60% [54].

The LIBRE study (Lifestyle Intervention in BRCA1/2 PV carriers) aims to randomise healthy BRCA1/2 PV carriers to a number of lifestyles interventions versus a control group and assess changes in physical and psychological well being [55].

13. Discussion

The worldwide burden of breast cancer poses significant challenges to health care providers. There is an increasing awareness that this heterogenous disease requires a multi-modality approach with consideration to both prevention and treatment.

Diet and lifestyle are important modifiers in the prevention of breast cancer. Adherence to physical activity and alcohol guidelines may reduce the risk of developing breast cancer (pre and post menopausal), with weight control being more important in prevention in the post menopausal cohort [56]. Energy restriction, in particular intermittently is association with changes in breast gene expression and systemic metabolism [57]. Future studies will need to assess the mechanism and clinical significance in breast cancer pathogenesis.

Breast cancer metabolomics is an area of growing interest. A recent study was able to determine a differential metabolic signature based on BRCA1 functionality [58] and may provide future biomarkers.

An area of interest is establishing whether breast cancer may be considered a communicable disease. There is established causality of infection with the Human Papilloma Virus and a number of cancers namely cervical cancer and associations with penile, anal and vulvar cancer. A similar aetiology has been postulated with breast cancer with entry of the virus via the nipple areolar complex resulting in biological activity within the mammary duct epithelium resulting in breast cancer. Data from Norway [59] have identified a possible association of pre-malignant changes in the cervix and an increased risk of subsequent breast cancer. A subsequent UK study [60] has identified the presence of high-risk HPV DNA in 42% of breast cancers with viral activity confirmed in less than 20% of invasive cancers suggesting that this may be an area of on going interest in the future.

Gene therapy in cancer aims at correcting specific genetic anomalies contributing to the development of certain cancers. This can be subdivided into germline and somatic cell gene therapy and is an important treatment modality in disease processes like cystic fibrosis and blood disorders. Several BRCA1 retroviral vectors were assessed in Phase 2 clinical trials in ovarian cancer almost two decades ago [61] with some initial potential as a therapeutic modality. The lack of progress in viral vectors suggests that newer techniques such as gene editing may find a place.

The UK 100,000 Genome project completed recruitment in 2018. Breast cancer patients represent a significant proportion of the 85,000 patients either with a cancer or rare disease diagnosis. Whole genome sequencing has allowed rapid analysis of the entire genome in this patient subgroup and is already aiding clinicians in making therapeutic decision in breast cancer, although no new hereditary genes have yet been identified from this initiative.

14. Conclusion

Pathogenic variants in the BRCA1 and BRCA2 gene confer a substantial lifetime risk of developing breast cancer. These breast cancers display characteristic clinical and pathological features that are important in the clinical management of the disease.

Multiple strategies exist for healthy individuals harbouring PVs in these genes. This comprises surveillance using modern day radiology (mammography and MRI scans) and risk reducing strategies. The latter include bilateral risk-reducing mastectomy, bilateral salpingo-oophorectomy, chemoprevention and lifestyle modification.

References

  1. 1. World Cancer Research Fund. Breast Cancer Statistics. 2018. Available from: https://www.wcrf.org/dietandcancer/cancer-trends/breast-cancer-statistics
  2. 2. Cancer Research UK. Breast Cancer. 2019. Available from: https://www.cancerresearchuk.org/about-cancer/breast-cancer/about
  3. 3. Union for International Cancer Control. New Global Cancer Data: GLOBOCAN 2018. 2019. Available from: https://www.uicc.org/news/new-global-cancer-data-globocan-2018
  4. 4. van der Borden CL, Stoffers S, Lips EH, Wesseling J. Avoiding overtreatment of ductal carcinoma in situ. Trends in Cancer. 2019;5(7):391-393
  5. 5. Makki J. Diversity of breast carcinoma: Histological subtypes and clinical relevance. Clinical Medicine Insights: Pathology. 2015;8:23-31
  6. 6. Lakhani S, Ellis I, Schnitt S, Tan P, van de Vijver M. WHO Classification of Breast Tumors. Lyon: IARC; 2012
  7. 7. Love RR, Philips J. Oophorectomy for breast cancer: History revisited. JNCI: Journal of the National Cancer Institute. 2002;94(19):1433-1434
  8. 8. Murphy CG, Dickler MN. Endocrine resistance in hormone-responsive breast cancer: Mechanisms and therapeutic strategies. Endocrine-Related Cancer. 2016;23(8):R337-R352
  9. 9. Hudis CA. Trastuzumab—Mechanism of action and use in clinical practice. The New England Journal of Medicine. 2007;357(1):39-51
  10. 10. Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. Surgical Oncology Clinics of North America. 2018;27(1):95-120
  11. 11. Fisher B, Anderson S, Bryant J, Margolese RG, Deutsch M, Fisher ER, et al. Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. The New England Journal of Medicine. 2002;347(16):1233-1241
  12. 12. Marra A, Viale G, Curigliano G. Recent advances in triple negative breast cancer: The immunotherapy era. BMC Medicine. 2019;17(1):90
  13. 13. Lalloo F, Evans DG. Familial breast cancer. Clinical Genetics. 2012;82(2):105-114
  14. 14. Mavaddat N, Peock S, Frost D, Ellis S, Platte R, Fineberg E, et al. Cancer risks for BRCA1 and BRCA2 mutation carriers: Results from prospective analysis of EMBRACE. Journal of the National Cancer Institute. 2013;105(11):812-822
  15. 15. Claus EB, Risch N, Thompson WD. Genetic analysis of breast cancer in the cancer and steroid hormone study. American Journal of Human Genetics. 1991;48(2):232-242
  16. 16. Sherkow JS, Greely HT. The history of patenting genetic material. Annual Review of Genetics. 2015;49:161-182
  17. 17. Evans DG, Shenton A, Woodward E, Lalloo F, Howell A, Maher ER. Penetrance estimates for BRCA1 and BRCA2 based on genetic testing in a Clinical Cancer Genetics service setting: Risks of breast/ovarian cancer quoted should reflect the cancer burden in the family. BMC Cancer. 2008;8:155
  18. 18. Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Devilee P, et al. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. American Journal of Human Genetics. 1998;62(3):676-689
  19. 19. Kuchenbaecker KB, Hopper JL, Barnes DR, Phillips KA, Mooij TM, Roos-Blom MJ, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA. 2017;317(23):2402-2416
  20. 20. Da Silva L, Lakhani SR. Pathology of hereditary breast cancer. Modern Pathology. 2010;23(Suppl 2):S46-S51
  21. 21. Mavaddat N, Barrowdale D, Andrulis IL, Domchek SM, Eccles D, Nevanlinna H, et al. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: Results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiology, Biomarkers & Prevention. 2012;21(1):134-147
  22. 22. Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D, Collins N, et al. Localization of a breast cancer susceptibility gene, BRCA2, to chromosome 13q12-13. Science. 1994;265(5181):2088-2090
  23. 23. Zhang F, Ma J, Wu J, Ye L, Cai H, Xia B, et al. PALB2 links BRCA1 and BRCA2 in the DNA-damage response. Current Biology: CB. 2009;19(6):524-529
  24. 24. Yang RL, Mick R, Lee K, Graves HL, Nathanson KL, Domchek SM, et al. DCIS in BRCA1 and BRCA2 mutation carriers: Prevalence, phenotype, and expression of oncodrivers C-MET and HER3. Journal of Translational Medicine. 2015;13:335
  25. 25. Copson ER, Maishman TC, Tapper WJ, Cutress RI, Greville-Heygate S, Altman DG, et al. Germline BRCA mutation and outcome in young-onset breast cancer (POSH): A prospective cohort study. The Lancet Oncology. 2018;19(2):169-180
  26. 26. van den Broek AJ, Schmidt MK, van ‘t Veer LJ, Tollenaar RA, van Leeuwen FE. Worse breast cancer prognosis of BRCA1/BRCA2 mutation carriers: What's the evidence? A systematic review with meta-analysis. PLoS ONE. 2015;10(3):e0120189
  27. 27. Pierce LJ, Phillips KA, Griffith KA, Buys S, Gaffney DK, Moran MS, et al. Local therapy in BRCA1 and BRCA2 mutation carriers with operable breast cancer: Comparison of breast conservation and mastectomy. Breast Cancer Research and Treatment. 2010;121(2):389-398
  28. 28. Gao X, Fisher SG, Emami B. Risk of second primary cancer in the contralateral breast in women treated for early-stage breast cancer: A population-based study. International Journal of Radiation Oncology, Biology, Physics. 2003;56(4):1038-1045
  29. 29. Drooger J, Akdeniz D, Pignol JP, Koppert LB, McCool D, Seynaeve CM, et al. Adjuvant radiotherapy for primary breast cancer in BRCA1 and BRCA2 mutation carriers and risk of contralateral breast cancer with special attention to patients irradiated at younger age. Breast Cancer Research and Treatment. 2015;154(1):171-180
  30. 30. Tassone P, Tagliaferri P, Perricelli A, Blotta S, Quaresima B, Martelli ML, et al. BRCA1 expression modulates chemosensitivity of BRCA1-defective HCC1937 human breast cancer cells. British Journal of Cancer. 2003;88(8):1285-1291
  31. 31. Lord CJ, Tutt AN, Ashworth A. Synthetic lethality and cancer therapy: Lessons learned from the development of PARP inhibitors. Annual Review of Medicine. 2015;66:455-470
  32. 32. Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, 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
  33. 33. Robson M, Im SA, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. The New England Journal of Medicine. 2017;377(6):523-533
  34. 34. Lee A, Moon BI, Kim TH. BRCA1/BRCA2 pathogenic variant breast cancer: Treatment and prevention strategies. Annals of Laboratory Medicine. 2020;40(2):114-121
  35. 35. Lostumbo L, Carbine NE, Wallace J. Prophylactic mastectomy for the prevention of breast cancer. Cochrane Database of Systematic Reviews. 2010;11:CD002748
  36. 36. Meijers-Heijboer EJ, Verhoog LC, Brekelmans CT, Seynaeve C, Tilanus-Linthorst MM, Wagner A, et al. Presymptomatic DNA testing and prophylactic surgery in families with a BRCA1 or BRCA2 mutation. Lancet. 2000;355(9220):2015-2020
  37. 37. Evans DG, Baildam AD, Anderson E, Brain A, Shenton A, Vasen HF, et al. Risk reducing mastectomy: Outcomes in 10 European centres. Journal of Medical Genetics. 2009;46(4):254-258
  38. 38. Evans DG, Lalloo F, Ashcroft L, Shenton A, Clancy T, Baildam AD, et al. Uptake of risk-reducing surgery in unaffected women at high risk of breast and ovarian cancer is risk, age, and time dependent. Cancer Epidemiology, Biomarkers & Prevention. 2009;18(8):2318-2324
  39. 39. Den Heijer M, van Asperen CJ, Harris H, Nippert I, Schmidtke J, Bouhnik AD, et al. International variation in physicians' attitudes towards prophylactic mastectomy—Comparison between France, Germany, the Netherlands and the United Kingdom. European Journal of Cancer. 2013;49(13):2798-2805
  40. 40. Metcalfe KA, Birenbaum-Carmeli D, Lubinski J, Gronwald J, Lynch H, Moller P, et al. International variation in rates of uptake of preventive options in BRCA1 and BRCA2 mutation carriers. International Journal of Cancer. 2008;122(9):2017-2022
  41. 41. Metcalfe KA, Ghadirian P, Rosen B, Foulkes W, Kim-Sing C, Eisen A, et al. Variation in rates of uptake of preventive options by Canadian women carrying the BRCA1 or BRCA2 genetic mutation. Open Medicine. 2007;1(2):e92-e98
  42. 42. Liede A, Cai M, Crouter TF, Niepel D, Callaghan F, Evans DG. Risk-reducing mastectomy rates in the US: A closer examination of the Angelina Jolie effect. Breast Cancer Research and Treatment. 2018;171(2):435-442
  43. 43. Evans DG, Barwell J, Eccles DM, Collins A, Izatt L, Jacobs C, et al. The Angelina Jolie effect: How high celebrity profile can have a major impact on provision of cancer related services. Breast Cancer Research. 2014;16(5):442
  44. 44. Evans DG, Wisely J, Clancy T, Lalloo F, Wilson M, Johnson R, et al. Longer term effects of the Angelina Jolie effect: Increased risk-reducing mastectomy rates in BRCA carriers and other high-risk women. Breast Cancer Research. 2015;17:143
  45. 45. Domchek SM, Friebel TM, Singer CF, Evans DG, Lynch HT, Isaacs C, et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA. 2010;304(9):967-975
  46. 46. Rebbeck TR, Lynch HT, Neuhausen SL, Narod SA, Van’t Veer L, Garber JE, et al. Prophylactic oophorectomy in carriers of BRCA1 or BRCA2 mutations. The New England Journal of Medicine. 2002;346(21):1616-1622
  47. 47. Heemskerk-Gerritsen BA, Seynaeve C, van Asperen CJ, Ausems MG, Collee JM, van Doorn HC, et al. Breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: Revisiting the evidence for risk reduction. Journal of the National Cancer Institute. 2015;107(5)djv033:1-9
  48. 48. Howell A, Anderson AS, Clarke RB, Duffy SW, Evans DG, Garcia-Closas M, et al. Risk determination and prevention of breast cancer. Breast Cancer Research. 2014;16(5):446
  49. 49. Taylor R, Taguchi K. Tamoxifen for breast cancer chemoprevention: Low uptake by high-risk women after evaluation of a breast lump. Annals of Family Medicine. 2005;3(3):242-247
  50. 50. Vogel VG, Costantino JP, Wickerham DL, Cronin WM, Cecchini RS, Atkins JN, et al. Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: The NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295(23):2727-2741
  51. 51. Goss PE, Ingle JN, Ales-Martinez JE, Cheung AM, Chlebowski RT, Wactawski-Wende J, et al. Exemestane for breast-cancer prevention in postmenopausal women. The New England Journal of Medicine. 2011;364(25):2381-2391
  52. 52. Cuzick J, Sestak I, Baum M, Buzdar A, Howell A, Dowsett M, et al. Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 10-year analysis of the ATAC trial. The Lancet Oncology. 2010;11(12):1135-1141
  53. 53. Parkin DM, Boyd L, Walker LC. 16. The fraction of cancer attributable to lifestyle and environmental factors in the UK in 2010. British Journal of Cancer. 2011;105(Suppl 2):S77-S81
  54. 54. Cloud AJ, Thai A, Liao Y, Terry MB. The impact of cancer prevention guideline adherence on overall mortality in a high-risk cohort of women from the New York site of the Breast Cancer Family Registry. Breast Cancer Research and Treatment. 2015;149(2):537-546
  55. 55. Kiechle M, Engel C, Berling A, Hebestreit K, Bischoff SC, Dukatz R, et al. Effects of lifestyle intervention in BRCA1/2 mutation carriers on nutrition, BMI, and physical fitness (LIBRE study): Study protocol for a randomized controlled trial. Trials. 2016;17:368
  56. 56. Harvie M, Howell A, Evans DG. Can diet and lifestyle prevent breast cancer: What is the evidence? American Society of Clinical Oncology Educational Book. 2015;35:e66-e73
  57. 57. Harvie MN, Sims AH, Pegington M, Spence K, Mitchell A, Vaughan AA, et al. Intermittent energy restriction induces changes in breast gene expression and systemic metabolism. Breast Cancer Research. 2016;18(1):57
  58. 58. Roig B, Rodriguez-Balada M, Samino S, Lam EW, Guaita-Esteruelas S, Gomes AR, et al. Metabolomics reveals novel blood plasma biomarkers associated to the BRCA1-mutated phenotype of human breast cancer. Scientific Reports. 2017;7(1):17831
  59. 59. Hansen BT, Nygard M, Falk RS, Hofvind S. Breast cancer and ductal carcinoma in situ among women with prior squamous or glandular precancer in the cervix: A register-based study. British Journal of Cancer. 2012;107(9):1451-1453
  60. 60. Salman NA, Davies G, Majidy F, Shakir F, Akinrinade H, Perumal D, et al. Association of high risk human papillomavirus and breast cancer: A UK based study. Scientific Reports. 2017;7:43591
  61. 61. Obermiller PS, Tait DL, Holt JT. Gene therapy for carcinoma of the breast: Therapeutic genetic correction strategies. Breast Cancer Research. 2000;2(1):28-31

Written By

Naren Basu and D. Gareth Evans

Submitted: 14 June 2019 Reviewed: 20 December 2019 Published: 20 January 2020