Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 24.
Published in final edited form as: Breast Cancer Res Treat. 2018 Sep 28;173(1):37–48. doi: 10.1007/s10549-018-4921-9

Male breast cancer: a disease distinct from female breast cancer

Ayca Gucalp a, Tiffany A Traina a, Joel R Eisner b, Joel S Parker c,d, Sara R Selitsky d, Ben H Park e, Anthony D Elias f, Edwina S Baskin-Bey b, Fatima Cardoso g
PMCID: PMC7513797  NIHMSID: NIHMS1623614  PMID: 30267249

Introduction

Male breast cancer (BC) is rare, representing approximately 1% of cancers that occur in men and approximately 1% of all BCs worldwide [16]. Less than 0.2% of cancer-related deaths in men can be attributed to male BC [7, 8]. Because male BC occurs at a very low incidence, BC literature, research, clinical trials, and development of new treatment options primarily focus on female BC. Although knowledge about female BC can inform male BC diagnosis and treatment, molecular and clinicopathologic features differ between male and female BC. Biologic factors, such as sex differences, hormonal regulation, and response to treatment (both tolerability and activity), must be considered when defining this disease in men and deciding upon treatment options [8]. Currently, no standard of care (SOC) exists for male BC, and there is an unmet need for research and therapeutic options for this disease. This review summarizes current knowledge of male BC to date with a discussion of future treatment options for the disease.

Demographics of Male Breast Cancer

Almost 1% of all BCs occur in men. In 2018, about 2550 men will be diagnosed with BC in the US, and male BC will account for 480 deaths. In comparison, it is estimated that there will be approximately 266,120 new cases of female BC and approximately 40,920 deaths due to BC in women [9, 10]. According to Surveillance, Epidemiology, and End Results Program (SEER) data from 2015, the incidence of invasive BC in men is 1.1:100,000 men, whereas it is 126.5:100,000 in women [11]. However, the incidence and prevalence of male BC varies by race and ethnicity, and men in certain select populations such as African Americans have higher rates of male BC in comparison to their Caucasian, Hispanic, or Asian/Pacific Islander counterparts [12]. The median age at initial diagnosis of invasive BC is typically older in men than in women (68 versus 62 years) [13]. In both men and women, the age-adjusted rate of BC sharply increases by the fifth decade of life. However, the rate plateaus in women by the sixth decade of life but continues to increase in men through the seventh decade.

Disease Characteristics of Male Breast Cancer

Initial diagnosis of male BC often occurs at a later stage than in female BC, and male BC often exhibits more advanced disease features, such as a larger tumor size, lymph node involvement, and distant metastases at the time of diagnosis [1, 2, 8, 1418]. In developed countries, nearly two-thirds of invasive female BCs are localized at the time of diagnosis; in contrast, in men approximately half of the cases are localized and the other half are regional or distant disease [11].

Estimates of in situ carcinoma in men are approximately 10%; the remaining 90% can be attributed to infiltrating ductal carcinoma [1, 2, 4, 8, 19]. Infiltrating lobular carcinoma, medullary lesions, and tubular or neuroendocrine tumors are very rare in men, as is triple-negative BC (TNBC) [1, 2, 4, 19, 20]. Male BCs usually express the estrogen receptor (ER), progesterone receptor (PR), and androgen receptor (AR); are hormonally responsive; and most commonly present as unilateral tumors [5, 1719, 2123]. Nipple retraction or the palpation of a retroareolar mass is a common physical examination finding and may be the first clinical manifestation of male BC [2].

Genetic/Epigenetic Alterations in Male Breast Cancer

A study by Johansson and colleagues revealed that there are 2 main subtypes of male BC: the male complex subtype, which is similar to female luminal BC, and the male simplex subtype, which does not exist in female BC [24, 25]. These findings have been confirmed by results from the International Male Breast Cancer Program (IMBCP) [20]. The IMBCP results also revealed additional important alterations using RNA sequencing.

In addition, the research laboratory of one of the authors (J. Parker) has been investigating the gene expression patterns of BC using RNA sequencing and other methods [26]. In the current analysis, gene expression, somatic mutation, and copy number data for 1098 BC samples were acquired from The Cancer Genome Atlas (TCGA) Firehose (The Broad Institute Genome Data Analysis Center, https://gdac.broadinstitute.org/). Sex information was abstracted from the clinical data for 451 samples, including 5 samples from men. Samples without sex information in the available clinical data were assigned as ‘likely’ male (N = 7) or female based on the expression of UTY. Gene expression intrinsic subtypes were assigned based on the PAM50 algorithm [27].

At the global level, BC gene expression analyzed with principal component analysis provides evidence that male BC is generally similar to female BC, as top components of variation do not segregate by sex. Instead, the predominant variation segregates by intrinsic subtype, with male BC overlapping with female luminal cases. Supporting this, the male BC samples were significantly enriched with luminal cells (Fisher’s Exact P = 0.009). This evidence that a majority of male BC samples tested were luminal-type is a distinctly different feature compared to female BC, which shows a greater diversity of subtypes. No difference in mutation rate was observed between female and male samples. Similar to female luminal tumors, individual mutations of GATA3 and PIK3CA are frequent, although there are no TP53 mutations in the male BC samples. In a recently published study, the most frequently mutated genes in male BC were PIK3CA (20%) and GATA3 (15%) [28], which agrees with the findings from the Parker laboratory.

From the perspective of copy number, differences in males and females are not observed after accounting for PAM50 intrinsic subtyping. While the number of males in the Parker dataset is small (N = 12), these data provide strong support that many if not most male BC will be characterized as luminal breast cancer. The characterization of male BC as luminal type is in agreement with recent studies in which 29% and 71% of male BCs were classified as luminal A–like and B–like, respectively, by immunohistochemistry and in which genetic tests of male BC samples showed a pattern of aberrations similar to female BC samples of a luminal subgroup [25, 28].

Although male BC seems globally similar to female BC, the evidence from studies summarized above indicates that there are common genetic features of female BC that are not shared with male BC. Underlying genetic and epigenetic differences between male and female BC exist and are summarized in Table 1. For male BC—as with many cancers—family history plays a large role in an individual’s risk of acquiring the disease. A family history of BC increases the risk of male BC [29] (a relative risk of 2.5 [4]), and 20% of men with BC have a first-degree relative with BC [6, 29]; the BC risk increases to more than 5-fold when the number of relatives with BC, especially early onset, increases [30]. Inherited germline mutations are a likely etiology for 4% to 40% of male BCs (versus 30% to 86% of female BCs) [6, 31, 32]. In particular, mutations in BRCA1 and especially BRCA2 are associated with increased BC risk [30]. Estimates of the lifetime risk of developing male BC range from 1% to 5% for BRCA1 mutation carriers and 5% to10 % for BRCA2 mutation carriers, compared to 0.1% in the general population [33].

Table 1.

Comparison of Male and Female Invasive Breast Cancer

Characteristic Male Female Comments
Patient Demographics
Incidence 2550 266,120 2018 US estimates [9, 10]
Annual deaths 480 40,920 2018 US estimates [9, 10]
Median age at diagnosis, years 68 [13] 62 [13]
Age-adjusted incidence rates (per 100,000 individuals)
 All years 1.2 124.8 SEER 2011–2015 [11]
 20–49 y 0.2 7.14
 50–64 y 1.8 270.1
 65–74 y 5.2 426.0
 75+ y 7.7 414.3
Disease Characteristics
BC distribution at diagnosis
 Localized 63.1% 45.4% SEER 2006–2015 [11]
 Regional 29.1% 43.6%
 Distant 5.7% 8.1%
 Unstaged 2.1% 2.9%
Histological type
 Ductal 89% 73% Prospective analysis of 449 male BC patients samples [44]; Retrospective analysis of 190,458 female BC cases [45]
 Lobular 1% 8%
 Other 9% 19%
Receptor status
 ER+ 99% 77% Retrospective analysis of 1483 male BC patients samples [20]; Retrospective analysis 2171 female BC patient samples [46]
 PR + 82% 64%
 AR+ 97% 77%
 HER2 + 9% 11%
 TNBC 0.3% 11%
Genetic/Epigenetic Alterations
CYP17 gene aberration Common Rare [36, 37]
Klinefelter’s syndrome (XXY) BC rates increase 20- to 50-fold compared to XY males [4] None [4]
Hypermethylation of BRCA1, BRCA2, CD44, ESR1, STK11, RARB, and ATM promoter regions Rare [39] Common [39]
BRCA1 germline mutation Rare (∼1%) [29] Rare ∼5–10%) [47]
BRCA2 germline mutation Common (∼12%) (60% – 76% in male BC patients with multiple family members with BC) [29]; pathogenic variants increase risk 13.9-fold [48] Rare (∼5%) [47]
CHEK2 mutations Pathogenic variants increase risk 3.7-fold [48]
CHEK2 1100delC deletion Deletion increases risk 3.13-fold [49] Deletion increases risk 2.88-fold [49]
PALB2 mutations Pathogenic variants increase risk 6.6-fold [48]

AR androgen receptor, ATM ataxia telangiectasia mutated, BC breast cancer, BRCA breast cancer gene, CD44 CD44 molecule (Indian blood group), CHEK2 checkpoint kinase 2, ER estrogen receptor, ESR1 estrogen receptor 1, HER2 human epidermal growth factor receptor 2, PALB2 partner and localizer of BRCA2, PR progesterone receptor, RARB retinoic acid receptor beta, SEER Surveillance, Epidemiology, and End Results Program, STK11 serine/threonine kinase 11, TNBC triple-negative breast cancer, US United States

Mutations in other DNA-repair genes such as CHEK2 [34] and PALB2 [35] are also associated with male BC. In a recently reported study of male BC patients undergoing testing with a multigene panel of 8 or more common cancer-related mutations, it was shown that more than 13.3% of the men tested positive for one or more of the mutations, and the most common mutations in that subset of patients were in BRCA2 (47%), followed by CHEK2 (31%), PALB2 (7%), BRCA1 (9%), and ATM (4%) [36]. In addition, genetic variation in CYP17, the gene that encodes an enzyme responsible for sex steroid production, has been proposed as a risk factor for breast and prostate cancer. Two case-control studies have investigated single nucleotide polymorphisms (SNPs) in CYP17 [37, 38]. One study found a polymorphism (a T to C substitution in the promoter region) associated with increased risk of male BC but not female BC [37], while the other study found a statistically nonsignificant increased risk of the same polymorphism in men with the BRCA2 999del5 mutation [38]. Another study of CYP17 in a large study of 8138 prostate cancer cases and 5333 BC cases from the Breast and Prostate Cancer Cohort Consortium found no association between common genetic variants in CYP17and prostate cancer or BC [39]. However, the majority of BC patients were female, and the results did not address male BC specifically.

Epigenetic alterations have also been implicated in the development and progression of cancer. Similar to female BC, promoter hypermethylation is also common in male BC, and the methylated genes are common between men and women [40]. The difference is that many of the genes investigated have decreased methylation frequency compared to female BC, which provides additional differences between male and female BC development and progression.

Endocrine Risk Factors Associated with Male Breast Cancer

Klinefelter’s syndrome, a rare genetic condition characterized by the presence of an extra X chromosome (XXY genotype) and associated with testicular dysgenesis, gynecomastia, an altered balance of androgens and estrogens, and increased gonadotrophins, increases the risk of developing male BC up to 50-fold compared to unaffected men [4, 41]. Similar to the increased estrogen levels associated with Klinefelter’s syndrome, the use of exogenous estrogens, such as estrogen treatment of prostate cancer [42] and hormone therapy for male-to-female transsexuals [43], increases the risk of male BC. Endogenous causes of hyperestrogenization in men, including obesity, cirrhosis, mumps orchitis, undescended testes, or testicular injury also have been associated with increased risk of male BC[4, 29]. In a comprehensive overview of risk factors for male BC, alcohol consumption, occupation, and radiation exposure were implicated as additional risk factors [4]. Interestingly, invasive lobular BC is very rare in men due to a lack of terminal breast lobules, except for those who are exposed to increased estrogens from endogenous or exogenous sources [44].

Table 1 presents a summary of the major differences between male and female invasive breast cancer.

Treatment Options for Male Breast Cancer

While significant efforts have been made to increase female BC awareness, screening, diagnostics, and treatment options, research and outreach directed at male BC has been limited until recently. Management of male BC has mainly relied on adopting clinical practices developed to treat female BC patients [2, 51]. Several studies have indicated that the overall survival (OS) estimates for male BC are significantly lower than for female BC [15, 21, 52], which emphasizes the need for earlier screening and effective treatments in male BC. However, it is important to note that many men with BC tend to have additional comorbidities and/or other neoplasms (e.g., prostate cancer, colon cancer, lung cancer) and are more likely to die from other causes compared to women with BC [53]. Because of this, some researchers suggest that disease-specific survival (DSS) may be a more accurate benchmark for male BC prognosis and survival rates than OS [8, 15] and in particular adjusted breast cancer–specific survival (adjusted for age, comorbidities, and stage at diagnosis). Studies have found that men had slightly better or equivalent DSS compared to women [53, 54], although note that these studies were not controlled for BC subtype. Importantly, both the retrospective joint analyses and the prospective registry parts of the IMBCP have shown that overall quality of care for male patients with breast cancer is worrisomely inferior than that for female patients with BC [19, 20].

Treatment Options for Early-stage Male Breast Cancer

Treatment for early-stage male BC includes 4 main treatment modalities: surgery, radiation therapy, chemotherapy, and endocrine therapy [1, 8, 55]. Typically, men with BC are treated with modified radical mastectomy, with axillary lymph node dissection or sentinel node biopsy [56]. Breast conservation or nipple-sparing or skin-sparing mastectomies may also be performed in selected cases; oncoplastic techniques should be used in view of the significant psychological and emotional impact of the physical consequences of locoregional therapies in male patients. Men are more likely than women to undergo mastectomy and to receive adjuvant radiotherapy as they are often diagnosed at a later stage and have nipple or skin involvement at diagnosis [29]. While there are limited data on chemotherapy use for male BC, clinicians choosing to use chemotherapy typically assess similar clinicopathologic risk factors (including tumor size, nodal involvement, hormone receptor status, HER2 status, and the underlying biology of the cancer) in male BC patients as they do in female BC patients with early-stage disease.

As a majority of male BCs express the ER, the use of endocrine therapy such as tamoxifen is routine for the management of male BC. Tamoxifen is the SOC for adjuvant endocrine therapy, and aromatase inhibitors should not be used alone in this setting. Aromatase inhibitors are not commonly used for the initial treatment of male BC. In gonad-intact men, aromatase inhibitors may cause a partial decrease in estrogens but also cause an increase in androgens, presumably from a lack of estradiol negative feedback on gonadotropin release [57, 58]. This increase in androgens could be problematic given the near-universal AR positivity of male BC. In addition, estrogen production in men is 80% by peripheral aromatization but 20% by direct testicular production, the latter not being inhibited by an aromatase inhibitor alone [insert REF post QC]. For these reasons, if an aromatase inhibitor is used, it is recommended that it be co-administered with chemical or surgical castration [1]. However, this combination has substantial side effects, which usually are associated with low compliance and should be reserved for very select cases. Tamoxifen, a selective estrogen receptor modulator therapy, is the current endocrine therapy of choice for male BC, given that nearly all male BC tumors are ER+ and that tamoxifen has been shown to improve survival rates in ER+ female BC [4, 8]. Although tamoxifen has been shown to be effective for male BC, it has drawbacks: frequency of recurrence and side effects that include hot flashes, changes in vision, cognitive changes, and a lower sex drive [51, 5962]. Such quality of life issues may discourage male patients from continuing treatment, as 20% to 25% of men with BC discontinue tamoxifen due to side effects [4, 62]. Retrospective observational analysis has shown that survival of men with ER+, lymph node–negative tumors improved with tamoxifen but not with aromatase inhibitors [63]. In male subjects with hormone receptor–positive (HR+) disease receiving adjuvant hormonal therapy, a significantly (P = 0.007) higher proportion of subjects who received aromatase inhibitors died (32%) compared to those who received tamoxifen (18%) [64], providing further support of tamoxifen in the adjuvant setting. A prospective, randomized, Phase 2 trial of tamoxifen ± gonadotropin-releasing hormone (GnRH) analog versus exemestane + GnRH analog investigating endocrine changes in men with HR+ BC found that tamoxifen or exemestane given in combination with GnRH analog resulted in reductions in sex steroids (testosterone and estradiol) and gonadotropins (follicle stimulating hormone and luteinizing hormone) while tamoxifen alone resulted in increases in sex steroid and gonadotropin concentrations that were significantly different than either combination [65]. Tumor assessments were not reported.

Treatment Options for Metastatic Male Breast Cancer

Since male BC is almost always ER+, the preferred treatment option for first-line therapy of metastatic disease is endocrine therapy [66]. Tamoxifen is again the treatment of choice, unless relapse occurs while on treatment with this agent. In this circumstance, other therapeutic options should be considered such as an aromatase inhibitor (preferably associated with a luteinizing hormone-releasing hormone agonist) or fulvestrant. Chemotherapy should be reserved for highly symptomatic or visceral crisis situations [66].

The evidence of activity of aromatase inhibitors or fulvestrant in metastatic or locally advanced male BC is primarily derived from case reports. In a series of 15 cases of patients with metastatic or locally advanced male BC who received aromatase inhibitors, 2 patients (13%) had a complete response, 4 patients (27%) had a partial response, 2 patients (13%) had stable disease, and 7 patients (47%) had progressive disease; the median progression-free survival (PFS) was 4.4 months [67]. A limited number of case studies of the selective estrogen receptor degrader fulvestrant in men with metastatic BC have shown extended partial responses or stable disease [6870]. Combinations of endocrine and targeted agents, such as mTOR and CDK inhibitors, can be used in metastatic male BC patients, as the same indications are used for their female counterparts. When chemotherapy is indicated, the same agents and regimens recommended for female metastatic BC should be used for male metastatic BC.

Clinical Trials in Male Breast Cancer

Due to the rarity of male BC, therapeutic clinical trials targeting solely male BC patients or larger BC trials with distinct male cohorts are limited. To date, most male BC patients treated in clinical trials have been enrolled in trials that predominantly enroll female patients but also allow enrollment of men when eligible. A report in 2010 indicated that there was only one therapeutic BC clinical trial that focused solely on men, which closed without patient enrollment due to logistical problems and pushback from trial sites [1]. Since then, there have been two additional interventional clinical trials opened with a focus on male BC. An overview of current or recent clinical trials in male BC is provided in Table 2. Clinical trials are organized by intervention, with a focus on male BC (i.e., solely male BC subjects or separate male BC cohort if enrolling female BC subjects), observational male BC, or interventional that specifically mention combined enrollment of male and female subjects.

Table 2.

Clinical Trials for Male Breast Cancer or Clinical Trials in Breast Cancer Enrolling Men

NCT Number / Study Type Status Population Total N (Male BC N) Arms Primary Endpoint Sponsor
Interventional Trial – Male BC Focus
NCT01638247 / open-label Phase 3 Ongoing, not recruiting Men only, hormone receptor +, 56 (56) Tamoxifen, tamoxifen + GnRH analogue, exemestane + GnRH analogue Estradiol blood concentration German Breast Group
NCT00217659 / open-label Phase 2 Withdrawn due to low accrual and site logistics [1] Men only, recurrent or metastatic or recurrent BC, ER+ or PR+ 0 Anastrozole + goserelin acetate PFS, OS, ORR Southwest Oncology Group
NCT02580448 / open-label Phase 1/2 Actively recruiting Men and postmenopausal women; Phase 1: women only, TNBC or ER+ / Phase 2: TNBC, AR+ or ER+ 175 (21) Seviteronel CBR16 (female TNBC and male BC), CBR24 (female ER+) Innocrin Pharmaceuticals
Non-interventional – Male BC Focus
NCT01703520 / Register-based, case-control Completed Nov 2017 Men only with or without male BC, > 35 yrs old, finasteride users or non-users 575,216 (575,216) N/A Association between finasteride exposure and the development of breast cancer in men. Merck Sharp & Dohme Corp.
NCT01101425 / observational Ongoing, not recruiting Men only, invasive breast carcinoma
Part 1: retrospective observational diagnosed since 1990–2010
Part 2: prospective observational recently (≤ 3 months) diagnosed
Not available N/A Survival, PFS, time to locoregional or distant relapse, time to second primary, treatment patterns, patient and disease characteristics, and biological characterization European Organisation for Research and Treatment of Cancer – EORTC; Translational Breast Cancer Research Consortium – TBCRC
Interventional Trial – Combined male and female BC cohort
NCT02437318 / placebo-controlled Phase 3 Ongoing, not recruiting Men and postmenopausal women, ER+, HER2−, identified PIK3CA status 572 Alpelisib + fulvestrant, placebo + fulvestrant PFS for patients with PIK3CA mutant status Novartis Pharmaceuticals
NCT02941926 / open-label Phase 3 Actively recruiting Men and women, recurrent or metastatic BC, ER+, PR+, HER2− 3775 Ribociclib + letrozole Safety and tolerability (AEs/SAEs) Novartis Pharmaceuticals
NCT00754325 / open-label Phase 2 Completed Jan 2014 Men (1) and postmenopausal women (99), ER+ and/or PR+ 100 Dasatinib + fulvestrant, fulvestrant PD or death, PFS Bristol-Myers Squibb
NCT00875979 / open-label Phase 1b/2 Completed Aug 2011 Men (1) and women (66), HER2+, locally advanced or metastatic BC 67 Trastuzumab emtansine 3 mg/kg + pertuzumab 420 mg, trastuzumab emtansine 3.6 mg/kg + pertuzumab 420 mg Objective response Hoffmann-La Roche
NCT02753595 / open-label Phase 1/2 Ongoing, not recruiting Men and women, metastatic BC, HER2− 114 PEGPH20 + eribulin (several doses) / eribulin RP2D and ORR Eisai Inc.
NCT02980341 / open-label Phase 1/2 Actively recruiting Men and women, advanced or metastatic BC, HER3+ 80 Drug U3-1402 Safety and tolerability (AEs/SAEs) and tumor response Daiichi Sankyo Co., Ltd.
NCT02060253 / open-label Phase 1 Completed June 2018 Men and women, advanced or metastatic BC, HER2+ 9 Ganetespib, paclitaxel, and trastuzumab with pertuzumab MTD and RP2D Memorial Sloan Kettering Cancer Center

AE adverse event, AR androgen receptor, Aug August, BC breast cancer, CBR16 clinical benefit rate at 16 weeks, CBR24 clinical benefit rate at 24 weeks, DLT dose-limiting toxicity, ER estrogen receptor, GnRH gonadotropin-releasing hormone, HER2 human epidermal growth factor receptor 2, HER3 human epidermal growth factor 3, Jan January, MTD maximum tolerated dose, N/A not applicable, ORR objective response rate, OS overall survival, PD progressive disease, PEGPH20 PEGylated recombinant human hyaluronidase, PFS progression-free survival, PR progesterone receptor, RP2D recommended Phase 2 dose, SAE serious adverse event, TEAE treatment-emergent adverse event, TNBC triple-negative breast cancer

Note: Bolded NCT numbers indicate clinical trials with only male participants.

ClinicalTrials.gov was searched for interventional or noninterventional BC clinical trials that are recruiting, enrolling by invitation, active, or completed. Of the more than 600 BC clinical trials listed, approximately one-third were not exclusionary to male participation. Overall, approximately 2% (N = 12) of the trials listed specifically mention the enrollment of men. A blog post in May 2016 indicated that the male BC clinical trial landscape is mostly the same as in 2013: male participants are eligible for only one-third of BC trials and about 18% of Phase 3 trials [71]. Of the 12 trials listed in ClinicalTrials.gov that specifically mention the enrollment of men, only 3 are interventional clinical trials (Table 2). Due to the lack of interventional clinical trial data, data from observational or retrospective trials, case studies, or meta-analyses are leveraged to inform male BC. However, these approaches have drawbacks compared to interventional trials, emphasizing the need for randomized, controlled, interventional male BC trials. Such an approach requires focused and coordinated efforts to increase the feasibility of successful recruitment and completion. However, rare disease trials are difficult to operate. For this reason, male BC patients should not be excluded from any BC trial unless there is a strong biological reason to do so. This will allow for the generation of data on efficacy of new agents in this rare patient population.

Multinational Consortium Approach

Given the paucity of male BC subjects for prospective observational or interventional clinical trials, multinational consortium studies are warranted. A major advancement on this front is the ongoing International Male Breast Cancer Program, coordinated by the European Organisation for Research and Treatment of Cancer (EORTC) and the Translational Breast Cancer Research Consortium (TBCRC), under the umbrella of the Breast International Group (BIG) and the North American Breast Cancer Group (NABCG), funded by the Breast Cancer Research Foundation (BCRF). This program has 3 parts. Part 1 is a retrospective joint central analysis of 1822 men, and Part 2 is a 30-month prospective registry that has enrolled 557 subjects. Part 3, which involves prospective clinical trials, has yet to be initiated.

Part 1 showed low levels of breast-conserving surgery in M0 patients even though a majority (56%) of men had T1 tumors [20]. Despite >90% of the tumors being ER+, only 77% of patients used adjuvant endocrine therapy. While half of the node-positive patients received adjuvant radiotherapy, 36% of N1 and 15% of N2 patients did not. Almost one-third of patients received adjuvant therapy. Centralized pathology review of these cases showed that the luminal A–like subtype was the most common [72], and that unlike female BC, histological grade was not associated with overall survival. An analysis of male BC precursor lesions found that ductal carcinoma in situ was the most commonly observed precursor lesion in male BC, and its presence seems to be associated with a better outcome, particularly in luminal B HER2+ cases [73].

Initial baseline data from 557 patients enrolled between 2013 and 2017 in the prospective registry (Part 2) were recently reported [45]. There continues to be discordance with normal treatment patterns with female BC: while 46% of M0 patients enrolled were node-positive, only 29% received radiotherapy, and while 98% of men had ER+ disease, only 65% received endocrine therapy. Longer follow-up, disease characterization, and comparison to a similar female BC population are ongoing.

To date, this program has helped to better understand male BC treatment patterns, tumor characteristics, and quality of life, while establishing the foundation for larger multinational therapeutic clinical trials, which is the goal for Part 3.

Future Perspectives on Male Breast Cancer and Treatment Options

The research and available literature on male BC has expanded in the past few decades, and the progress of the IMBCP is an important step; however, knowledge is limited as clinical/translational research on male BC is still in its infancy. Although common female BC treatment practices are applied to the management of male BC, the medical community recognizes male BC as a distinct disease characterized by varied molecular and clinicopathologic features. Modified radical mastectomy as well as radiotherapy and adjuvant tamoxifen are still used in the treatment of male BC; however, the role of adjuvant systemic therapies in male BC is less understood [8]. Several endocrine, cytotoxic, and targeted therapies studied in in the metastatic male BC setting have shown variable responses [1, 4, 8, 14, 22, 74]. As indicated in Table 2, some ongoing BC trials evaluating a variety of new therapies include male patient recruitment and will provide important information. However, research specifically dedicated to male BC, especially research related to the best endocrine-based approaches, is warranted.

As discussed above, DNA damage repair (DDR) genes, specifically BRCA2, are frequently mutated or overexpressed in male BC. Given this, therapies targeting the poly-ADP-ribose polymerase (PARP) family of proteins could provide therapeutic benefit to male BC patients and warrant investigation. Specifically, PARP1 has been the focus of the most oncology research, as it is involved in 80% to 90% of DNA repair via the base excision repair (BER) pathway [75, 76]. Inhibition of PARP causes accumulation of single-strand DNA breaks, which are converted to double-strand breaks during DNA replication [77, 78]. Since mutation of BRCA2 results in the inability to resolve double-strand breaks (DSBs) via the high-fidelity homologous recombination pathway, DSBs are resolved by the more error-prone nonhomologous end joining pathway, which accelerates genomic instability to facilitate tumor cell death via the principle of synthetic lethality [78]. Knowledge about the efficacy of PARP1 in male BC is very limited. However, PARP inhibitors have shown activity in other BRCA-mutated male tumors, including prostate cancer. Indeed, results from the TOPARP-A trial (Trial of PARP Inhibition in Prostate Cancer) showed 88% of the patients who had a DDR pathway mutation responded to the PARP inhibitor, olaparib [79]. Thus, investigation of PARP inhibitors in DDR-deficient male BC should be considered.

The role of AR in BC is an area of active research [80], and AR has become a promising potential target for the treatment of male BC [51]. Recently, AR has been found to have prognostic significance in ER+ male BC [81]. A review by Imamura and Sadar provide a list of current therapies targeting AR that are used to treat castration-resistant prostate cancer (CRPC) [82], and some of these therapies are also potential candidates for treating male BC. To date, data exists from female BC trials of 3 compounds that have activity in CRPC: bicalutamide (a competitive inhibitor of AR binding), enzalutamide (a competitive inhibitor of AR binding and signaling), and seviteronel (a CYP17 lyase and AR inhibitor). A trial of the third compound, sevitoronel, includes separate male BC activity data.

In a Phase 2 trial of bicalutamide in women with metastatic AR+ TNBC, 5 of 26 patients had stable disease for at least 6 months. The resulting 24-week clinical benefit rate (CBR, the proportion of patients who showed a complete response, partial response, or stable disease) was 19% [83], and the median PFS was 12 weeks (95% confidence interval [CI], 11–22 weeks). Bicalutamide was well tolerated in this patient population and could provide a potential new therapeutic option for male BC, but it has not been evaluated to date.

Similarly, the activity of enzalutamide has been suggested in AR+ TNBC and AR+/ER+ BC [84, 85]. For patients with AR+ metastatic TNBC, a Phase 2 study of single-agent enzalutamide 160 mg met its primary endpoint with a CBR at 16 weeks of 35% and a median PFS of 14 weeks. Eight percent of patients had either a partial or complete response confirmed by RECIST criteria [86]. A randomized, placebo-controlled, Phase 2 study was conducted to evaluate exemestane with or without enzalutamide in 247 patients with HR+/HER2- male BC. The study met its primary endpoint, improving median PFS with the addition of enzalutamide to exemestane in the prespecified subset of patients who tested positive for the predictive biomarker developed in parallel [87]. Comparable improvement in PFS was not observed in women who had 1 prior endocrine therapy for advanced BC. Enzalutamide was well tolerated in both of these studies [87, 88]. Despite initial activity in female BC patients, development of enzalutamide in BC and the planned Phase 3 trial in TNBC were suspended [89].

Seviteronel, a nonsteroidal CYP17A1 and AR inhibitor, is in Phase 2 development for advanced prostate and breast cancers (Table 2), and unlike bicalutamide or enzalutamide, it has specifically been investigated in male BC [82]. Seviteronel blocks the synthesis of sex steroids (androgens and downstream estrogens) via CYP17 lyase inhibition and competitively inhibits the AR [90]. Fast-track designation was granted for seviteronel for CRPC in 2016 [91] and for male and female BC in 2017 [92]. Full results for the initial Phase 2 trials of seviteronel in BC and CRPC are not yet available; however, preliminary data from the Phase 2 trial in BC (NCT02580448) indicate that clinical benefit in Stage 1 of the trial was encouraging and sufficient to support accrual to Stage 2 for both women with ER+ BC and women with TNBC [93] as well as men with BC. Early Phase 2 results for the male BC cohort recently reported that 2 of the first 7 subjects enrolled met the initial activity endpoint of CBR at 16 weeks, with 1 of 2 patients remaining on seviteronel at 24 weeks. The most common adverse event was fatigue [94]. Phase 2 enrollment is ongoing. While these results are preliminary, this is the largest prospective therapeutic male BC clinical trial to date and the results highlight the potential for AR-targeted agents for the treatment of male BC.

Conclusions

While male BC is a rare and often neglected disease, there is a growing understanding of the biological differences between male and female BC. Such differences indicate that male BC should be seen as a separate disease, distinct from female BC. The effective treatment of patients with male BC remains a clinically challenging scenario with many unanswered questions. Research in this area is focused on evaluating the underlying biology of this subtype of tumors with the goal of developing improved therapeutic strategies for the management of this underrepresented population. An understanding of germline mutations with increased prevalence in male BC, such as BRCA2, may help with the identification of novel treatment options such as PARP inhibitors. Also, given the near universal expression of the AR in male BC tumors and relatively benign safety profile, AR-targeted agents may prove beneficial as a treatment option, either as monotherapy or in combination with other agents. Early activity of seviteronel in male BC highlights the targeting of the sex-steroid biosynthesis pathway and AR activation as a promising therapeutic approach for male BC. Future research in this area will require coordinated multicenter international collaboration to conduct larger, successful therapeutic clinical trials in male BC that lead to the development and approval of novel targeted agents for the management of this rare disease.

Acknowledgments

The authors thank Kelly Kilibarda, PhD and Elise Eller, PhD of Whitsell Innovations, Inc., Chapel Hill, NC, USA for providing medical writing support, which was funded by Innocrin Pharmaceuticals Inc., Durham, North Carolina, USA, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3).

Disclosure of Potential Conflicts of Interest

Ayca Gucalp receives salary support to conduct clinical trials from Innocrin Pharmaceuticals, Pfizer, Novartis, and Merck. She also received an honorarium for participating on an advisory board for Pfizer. Tiffany A. Traina is a compensated Seviteronel Breast Cancer Steering Committee member (Innocrin Pharmaceuticals) and receives research support from Innocrin Pharmaceuticals for trials of seviteronel. Joel R. Eisner and Edwina S. Baskin-Bey declare that they are employed by and have stock ownership in Innocrin Pharmaceuticals. Joel S. Parker is a compensated advisor to Innocrin Pharmaceuticals and author of the PAM50 patent and related patents for Nanostring Technologies, Inc. Ben H. Park has ownership interest and is a paid member of the scientific advisory board of Loxo Oncology and is a paid consultant for Foundation Medicine, Inc.; Jackson Laboratories; Casdin Capital; and Roche. Under separate licensing agreements between Horizon Discovery, Ltd and The Johns Hopkins University, Ben H. Park is entitled to a share of royalties received by the university on sales of products. The terms of this arrangement are managed by The Johns Hopkins University in accordance with its conflict of interest policies. Fatima Cardoso serves on advisory boards for Amgen, Astellas/Medivation, AstraZeneca, Celgene, Daiichi-Sankyo, Eisai, GE Oncology, Genentech, GlaxoSmithKline, Macrogenics, Merck-Sharp, Merus BV, Mylan, Mundipharma, Novartis, Pfizer, Pierre-Fabre, Roche, Sanofi, Seattle-Genetics, and Teva. All other authors declare that they have no potential conflict of interest.

References

  • 1.Korde LA, Zujewski JA, Kamin L, Giordano S, Domchek S, Anderson WF et al. (2010) Multidisciplinary meeting on male breast cancer: summary and research recommendations. J Clin Oncol 28(12): 2114–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cutuli B (2007) Strategies in treating male breast cancer. Expert Opin Pharmacother 8(2): 193–202 [DOI] [PubMed] [Google Scholar]
  • 3.Gennari R, Curigliano G, Jereczek-Fossa BA, Zurrida S, Renne G, Intra M et al. (2004) Male breast cancer: a special therapeutic problem. Anything new? (Review). Int J Oncol 24(3): 663–70 [PubMed] [Google Scholar]
  • 4.Fentiman IS, Fourquet A, Hortobagyi GN (2006) Male breast cancer. Lancet 367(9510): 595–604 [DOI] [PubMed] [Google Scholar]
  • 5.Yoney A, Kucuk A, Unsal M (2009) Male breast cancer: a retrospective analysis. Cancer Radiother 13(2): 103–7 [DOI] [PubMed] [Google Scholar]
  • 6.Weiss JR, Moysich KB, Swede H (2005) Epidemiology of male breast cancer. Cancer Epidemiol Biomarkers Prev 14(1): 20–6 [PubMed] [Google Scholar]
  • 7.Burga AM, Fadare O, Lininger RA, Tavassoli FA (2006) Invasive carcinomas of the male breast: a morphologic study of the distribution of histologic subtypes and metastatic patterns in 778 cases. Virchows Arch 449(5): 507–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Darkeh MHSE, Azavedo E (2014) Male breast cancer clinical features, risk factors, and current diagnostic and therapeutic approaches. Int J Clin Med (5): 1068–86 [Google Scholar]
  • 9.Key statistics for breast cancer in men. [Accessed March 5, 2018] Available from: https://www.cancer.org/cancer/breast-cancer-in-men/about/key-statistics.html.
  • 10.How common is breast cancer? [Accessed March 5, 2018] Available from: https://www.cancer.org/cancer/breast-cancer/about/how-common-is-breast-cancer.html.
  • 11.SEER*Explorer: an interactive website for SEER cancer statistics [internet]. [Accessed July 16, 2018] Available from: https://seer.cancer.gov/explorer/
  • 12.Howlader N, Noone AM, Krapcho M, Miller D BK, Altekruse SF, Kosary CL et al. (2016) SEER cancer statistics review, 1975–2013. 2016, National Cancer Institute: Bethesda, MD [Google Scholar]
  • 13.Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL et al. (2017) SEER cancer statistics review, 1975–2014. 2017, National Cancer Institute: Bethesda, MD [Google Scholar]
  • 14.Giotta F, Acito L, Candeloro G, Del Medico P, Gadaleta-Caldarola G, Giordano G et al. (2016) Eribulin in male patients with breast cancer: the first report of clinical outcomes. Oncologist [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Donegan WL, Redlich PN, Lang PJ, Gall MT (1998) Carcinoma of the breast in males: a multiinstitutional survey. Cancer 83(3): 498–509 [DOI] [PubMed] [Google Scholar]
  • 16.Joshi MG, Lee AK, Loda M, Camus MG, Pedersen C, Heatley GJ et al. (1996) Male breast carcinoma: an evaluation of prognostic factors contributing to a poorer outcome. Cancer 77(3): 490–8 [DOI] [PubMed] [Google Scholar]
  • 17.Arslan UY, Oksuzoglu B, Ozdemir N, Aksoy S, Alkis N, Gok A et al. (2012) Outcome of non-metastatic male breast cancer: 118 patients. Med Oncol 29(2): 554–60 [DOI] [PubMed] [Google Scholar]
  • 18.Anderson WF, Jatoi I, Tse J, Rosenberg PS (2010) Male breast cancer: a population-based comparison with female breast cancer. J Clin Oncol 28(2): 232–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Giordano SH, Schröder CP, Poncet C, van Leeuwen-Stok E, Linderholm B, Abreu MH et al. (2018) Clinical and biological characterization of male breast cancer (BC) EORTC 10085/TBCRC 029/BOOG 2013–02/BIG 2–07: Baseline results from the prospective registry. Cancer Research 78(4 Supplement): P5–23-01 [Google Scholar]
  • 20.Cardoso F, Bartlett JMS, Slaets L, van Deurzen CHM, van Leeuwen-Stok E, Porter P et al. (2018) Characterization of male breast cancer: results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Ann Oncol 29(2): 405–417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Giordano SH, Cohen DS, Buzdar AU, Perkins G, Hortobagyi GN (2004) Breast carcinoma in men: a population-based study. Cancer 101(1): 51–7 [DOI] [PubMed] [Google Scholar]
  • 22.Ruddy KJ, Winer EP (2013) Male breast cancer: risk factors, biology, diagnosis, treatment, and survivorship. Ann Oncol 24(6): 1434–43 [DOI] [PubMed] [Google Scholar]
  • 23.Everson RB, Lippman ME, Thompson EB, McGuire WL, Wittliff JL, De Sombre ER et al. (1980) Clinical correlations of steroid receptors and male breast cancer. Cancer Res 40(4): 991–7 [PubMed] [Google Scholar]
  • 24.Johansson I, Nilsson C, Berglund P, Lauss M, Ringner M, Olsson H et al. (2012) Gene expression profiling of primary male breast cancers reveals two unique subgroups and identifies N-acetyltransferase-1 (NAT1) as a novel prognostic biomarker. Breast Cancer Res 14(1): R31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Johansson I, Nilsson C, Berglund P, Strand C, Jonsson G, Staaf J et al. (2011) High-resolution genomic profiling of male breast cancer reveals differences hidden behind the similarities with female breast cancer. Breast Cancer Res Treat 129(3): 747–60 [DOI] [PubMed] [Google Scholar]
  • 26.Parker JS, Peterson AC, Tudor IC, Hoffman J, Uppal H (2015) A novel biomarker to predict sensitivity to enzalutamide (ENZA) in TNBC. J Clin Oncol 33(15 Suppl): 1083–1083 [Google Scholar]
  • 27.Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T et al. (2009) Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 27(8): 1160–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Piscuoglio S, Ng CK, Murray MP, Guerini-Rocco E, Martelotto LG, Geyer FC et al. (2016) The genomic landscape of male breast cancers. Clin Cancer Res 22(16): 4045–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Rudlowski C (2008) Male breast cancer. Breast Care (Basel) 3(3): 183–189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Rizzolo P, Silvestri V, Tommasi S, Pinto R, Danza K, Falchetti M et al. (2013) Male breast cancer: genetics, epigenetics, and ethical aspects. Ann Oncol 24 Suppl 8: viii75–viii82 [DOI] [PubMed] [Google Scholar]
  • 31.Ford D, Easton DF, Stratton M, Narod S, Goldgar D, Devilee P et al. (1998) Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. Am J Hum Genet 62(3): 676–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Greene MH (1997) Genetics of breast cancer. Mayo Clin Proc 72(1): 54–65 [DOI] [PubMed] [Google Scholar]
  • 33.Silvestri V, Barrowdale D, Mulligan AM, Neuhausen SL, Fox S, Karlan BY et al. (2016) Male breast cancer in BRCA1 and BRCA2 mutation carriers: pathology data from the Consortium of Investigators of Modifiers of BRCA1/2. Breast Cancer Res 18(1): 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Falchetti M, Lupi R, Rizzolo P, Ceccarelli K, Zanna I, Calo V et al. (2008) BRCA1/BRCA2 rearrangements and CHEK2 common mutations are infrequent in Italian male breast cancer cases. Breast Cancer Res Treat 110(1): 161–7 [DOI] [PubMed] [Google Scholar]
  • 35.Silvestri V, Rizzolo P, Zanna I, Falchetti M, Masala G, Bianchi S et al. (2010) PALB2 mutations in male breast cancer: a population-based study in Central Italy. Breast Cancer Res Treat 122(1): 299–301 [DOI] [PubMed] [Google Scholar]
  • 36.Vogel Postula KJ, Andolina LM, Theobald K, McGill AK, Sutcliffe E, Arvai KJ et al. (2018) The role of multi-gene hereditary cancer panels in male patients with breast cancer. Cancer Res 78(4 Suppl): PD7–11 [Google Scholar]
  • 37.Young IE, Kurian KM, Annink C, Kunkler IH, Anderson VA, Cohen BB et al. (1999) A polymorphism in the CYP17 gene is associated with male breast cancer. Br J Cancer 81(1): 141–3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gudmundsdottir K, Thorlacius S, Jonasson JG, Sigfusson BF, Tryggvadottir L, Eyfjord JE (2003) CYP17 promoter polymorphism and breast cancer risk in males and females in relation to BRCA2 status. Br J Cancer 88(6): 933–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Setiawan VW, Schumacher FR, Haiman CA, Stram DO, Albanes D, Altshuler D et al. (2007) CYP17 genetic variation and risk of breast and prostate cancer from the National Cancer Institute Breast and Prostate Cancer Cohort Consortium (BPC3). Cancer Epidemiol Biomarkers Prev 16(11): 2237–46 [DOI] [PubMed] [Google Scholar]
  • 40.Kornegoor R, Moelans CB, Verschuur-Maes AH, Hogenes M, de Bruin PC, Oudejans JJ et al. (2012) Promoter hypermethylation in male breast cancer: analysis by multiplex ligation-dependent probe amplification. Breast Cancer Res 14(4): R101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hultborn R, Hanson C, Kopf I, Verbiene I, Warnhammar E, Weimarck A (1997) Prevalence of Klinefelter’s syndrome in male breast cancer patients. Anticancer Res 17(6D): 4293–7 [PubMed] [Google Scholar]
  • 42.McClure J, Higgins C (1951) Bilateral carcinoma of male breast after estrogen therapy. J Am Med Assoc 146(1): 7–9 [DOI] [PubMed] [Google Scholar]
  • 43.Symmers WS (1968) Carcinoma of breast in trans-sexual individuals after surgical and hormonal interference with the primary and secondary sex characteristics. Br Med J 2(5597): 83–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ottini L, Palli D, Rizzo S, Federico M, Bazan V, Russo A (2010) Male breast cancer. Crit Rev Oncol Hematol 73(2): 141–55 [DOI] [PubMed] [Google Scholar]
  • 45.Giordano S, Schroder C, Poncet C, van Leeuwen-Stok E, Linderholm B, Abreu MH et al. (2017) Clinical and biological characterization of male breast cancer (MBC) EORTC 10085/TBCRC023: baseline results from the prospective registry. Presented at the 2017 San Antonio Breast Cancer Symposium December 5–9, 2017: San Antonio, Texas, USA Poster P5–23-01 [Google Scholar]
  • 46.Li CI, Anderson BO, Daling JR, Moe RE (2003) Trends in incidence rates of invasive lobular and ductal breast carcinoma. JAMA 289(11): 1421–4 [DOI] [PubMed] [Google Scholar]
  • 47.Collins LC, Cole KS, Marotti JD, Hu R, Schnitt SJ, Tamimi RM (2011) Androgen receptor expression in breast cancer in relation to molecular phenotype: results from the Nurses’ Health Study. Mod Pathol 24(7): 924–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Karami F, Mehdipour P (2013) A comprehensive focus on global spectrum of BRCA1 and BRCA2 mutations in breast cancer. Biomed Res Int 2013: 928562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Pritzlaff M, Summerour P, McFarland R, Li S, Reineke P, Dolinsky JS et al. (2017) Male breast cancer in a multi-gene panel testing cohort: insights and unexpected results. Breast Cancer Res Treat 161(3): 575–586 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liang M, Zhang Y, Sun C, Rizeq FK, Min M, Shi T et al. (2018) Association between CHEK2*1100delC and breast cancer: a systematic review and meta-analysis. Mol Diagn Ther [DOI] [PubMed] [Google Scholar]
  • 51.Severson TM, Zwart W (2017) A review of estrogen receptor/androgen receptor genomics in male breast cancer. Endocr Relat Cancer 24(3): R27–R34 [DOI] [PubMed] [Google Scholar]
  • 52.Wu Q, Li J, Zhu S, Wu J, Li X, Liu Q et al. (2016) Poorer breast cancer survival outcomes in males than females might be attributable to tumor subtype. Oncotarget 7(52): 87532–87542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.El-Tamer MB, Komenaka IK, Troxel A, Li H, Joseph KA, Ditkoff BA et al. (2004) Men with breast cancer have better disease-specific survival than women. Arch Surg 139(10): 1079–82 [DOI] [PubMed] [Google Scholar]
  • 54.Marchal F, Salou M, Marchal C, Lesur A, Desandes E (2009) Men with breast cancer have same disease-specific and event-free survival as women. Ann Surg Oncol 16(4): 972–8 [DOI] [PubMed] [Google Scholar]
  • 55.Bagley CS, Wesley MN, Young RC, Lippman ME (1987) Adjuvant chemotherapy in males with cancer of the breast. Am J Clin Oncol 10(1): 55–60 [DOI] [PubMed] [Google Scholar]
  • 56.Scott-Conner CE, Jochimsen PR, Menck HR, Winchester DJ (1999) An analysis of male and female breast cancer treatment and survival among demographically identical pairs of patients. Surgery 126(4): 775–80; discussion 780–1 [PubMed] [Google Scholar]
  • 57.Mauras N, O’Brien KO, Klein KO, Hayes V (2000) Estrogen suppression in males: metabolic effects. J Clin Endocrinol Metab 85(7): 2370–7 [DOI] [PubMed] [Google Scholar]
  • 58.Trunet PF, Mueller P, Bhatnagar AS, Dickes I, Monnet G, White G (1993) Open dose-finding study of a new potent and selective nonsteroidal aromatase inhibitor, CGS 20 267, in healthy male subjects. J Clin Endocrinol Metab 77(2): 319–23 [DOI] [PubMed] [Google Scholar]
  • 59.Kiluk JV, Lee MC, Park CK, Meade T, Minton S, Harris E et al. (2011) Male breast cancer: management and follow-up recommendations. Breast J 17(5): 503–9 [DOI] [PubMed] [Google Scholar]
  • 60.Arnould N, Pouget O, Gharbi M, Brettes JP (2006) Breast cancer in men: are there similarities with breast cancer in women? Gynecol Obstet Fertil 34(5): 413–9 [DOI] [PubMed] [Google Scholar]
  • 61.Anelli TF, Anelli A, Tran KN, Lebwohl DE, Borgen PI (1994) Tamoxifen administration is associated with a high rate of treatment-limiting symptoms in male breast cancer patients. Cancer 74(1): 74–7 [DOI] [PubMed] [Google Scholar]
  • 62.Visram H, Kanji F, Dent SF (2010) Endocrine therapy for male breast cancer: rates of toxicity and adherence. Curr Oncol 17(5): 17–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Harlan LC, Zujewski JA, Goodman MT, Stevens JL (2010) Breast cancer in men in the United States: a population-based study of diagnosis, treatment, and survival. Cancer 116(15): 3558–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Eggemann H, Ignatov A, Smith BJ, Altmann U, von Minckwitz G, Rohl FW et al. (2013) Adjuvant therapy with tamoxifen compared to aromatase inhibitors for 257 male breast cancer patients. Breast Cancer Res Treat 137(2): 465–70 [DOI] [PubMed] [Google Scholar]
  • 65.Reinisch M, Seiler S, Hauzenberger T, Schmatloch S, Strittmatter HJ, Zahm DM et al. (2017) Male-GBG54: A prospective, randomised, multi-centre, phase II study evaluating endocrine treatment with either tamoxifen +/− gonadotropin releasing hormone analogue (GnRHa) or an aromatase inhibitor + GnRHa in male breast cancer patients. Presented at the 2017 San Antonio Breast Cancer Symposium December 5–9, 2017: San Antonio, Texas USA Poster PD7–10. [Google Scholar]
  • 66.Cardoso F, Costa A, Senkus E, Aapro M, Andre F, Barrios CH et al. (2017) 3rd ESO-ESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 3). Ann Oncol 28(12): 3111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Doyen J, Italiano A, Largillier R, Ferrero JM, Fontana X, Thyss A (2010) Aromatase inhibition in male breast cancer patients: biological and clinical implications. Ann Oncol 21(6): 1243–5 [DOI] [PubMed] [Google Scholar]
  • 68.Masci G, Gandini C, Zuradelli M, Pedrazzoli P, Torrisi R, Lutman FR et al. (2011) Fulvestrant for advanced male breast cancer patients: a case series. Ann Oncol 22(4): 985. [DOI] [PubMed] [Google Scholar]
  • 69.de la Haba Rodríguez JR, Porras Quintela I, Pulido Cortio G, Berciano Guerrero M, Aranda E (2009) Fulvestrant in advanced male breast cancer. Ann Oncol 20(11): 1896–1897 [DOI] [PubMed] [Google Scholar]
  • 70.Agrawal A, Ayantunde AA, Rampaul R, Robertson JF (2007) Male breast cancer: a review of clinical management. Breast Cancer Res Treat 103(1): 11–21 [DOI] [PubMed] [Google Scholar]
  • 71.Helwick C Is male breast cancer overlooked in clinical trials? May 25, 2016. [Accessed March 12, 2017] Available from: http://www.ascopost.com/issues/may-25-2016/is-male-breast-cancer-overlooked-in-clinical-trials/.
  • 72.Vermeulen MA, Slaets L, Cardoso F, Giordano SH, Tryfonidis K, van Diest PJ et al. (2017) Pathological characterisation of male breast cancer: Results of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Eur J Cancer 82: 219–227 [DOI] [PubMed] [Google Scholar]
  • 73.Doebar SC, Slaets L, Cardoso F, Giordano SH, Bartlett JM, Tryfonidis K et al. (2017) Male breast cancer precursor lesions: analysis of the EORTC 10085/TBCRC/BIG/NABCG International Male Breast Cancer Program. Mod Pathol 30(4): 509–518 [DOI] [PubMed] [Google Scholar]
  • 74.Di Lauro L, Pizzuti L, Barba M, Sergi D, Sperduti I, Mottolese M et al. (2015) Efficacy of chemotherapy in metastatic male breast cancer patients: a retrospective study. J Exp Clin Cancer Res 34: 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Papa A, Caruso D, Strudel M, Tomao S, Tomao F (2016) Update on Poly-ADP-ribose polymerase inhibition for ovarian cancer treatment. J Transl Med 14: 267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Stepnik M, Spryszynska S, Gorzkiewicz A, Ferlinska M (2017) Cytotoxicity of anticancer drugs and PJ-34 (poly(ADP-ribose)polymerase-1 (PARP-1) inhibitor) on HL-60 and Jurkat cells. Adv Clin Exp Med 26(3): 379–385 [DOI] [PubMed] [Google Scholar]
  • 77.Kim Y, Kim A, Sharip A, Sharip A, Jiang J, Yang Q et al. (2017) Reverse the resistance to PARP inhibitors. Int J Biol Sci 13(2): 198–208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.del Rivero J, Kohn EC (2017) PARP Inhibitors: The Cornerstone of DNA Repair-Targeted Therapies. Oncology (Williston Park) 31(4): 265–73 [PubMed] [Google Scholar]
  • 79.Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R et al. (2015) DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med 373(18): 1697–708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Christenson JL, Trepel JB, Haythem YA, Lee S, Eisner JR, Baskni-Bey ES et al. (2018) Harnessing a different dependency: how to identify and target androgen receptor-positive versus quadruple-negative breast cancer. Horm Canc 9(2): 82–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Humphries MP, Sundara Rajan S, Honarpisheh H, Cserni G, Dent J, Fulford L et al. (2017) Characterisation of male breast cancer: a descriptive biomarker study from a large patient series. Sci Rep 7: 45293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Imamura Y, Sadar MD (2016) Androgen receptor targeted therapies in castration-resistant prostate cancer: Bench to clinic. Int J Urol 23(8): 654–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gucalp A, Tolaney S, Isakoff SJ, Ingle JN, Liu MC, Carey LA et al. (2013) Phase II trial of bicalutamide in patients with androgen receptor-positive, estrogen receptor-negative metastatic Breast Cancer. Clin Cancer Res 19(19): 5505–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cochrane DR, Bernales S, Jacobsen BM, Cittelly DM, Howe EN, D’Amato NC et al. (2014) Role of the androgen receptor in breast cancer and preclinical analysis of enzalutamide. Breast Cancer Res 16(1): R7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rahim B, O’Regan R (2017) AR signaling in breast cancer. Cancers (Basel) 9(3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Traina TA, Miller K, Yardley DA, O’Shaughnessy J, Cortes J, Awada A et al. (2015) Results from a Phase 2 study of enzalutamide, an androgen receptor inhibitor, in advanced AR+ triple-negative breast cancer (MDV3100–11). Presented at the 2015 American Society of Clinical Oncology annual conference May 29-June 2, 2015: Chicago, Illinois USA [Google Scholar]
  • 87.Yardley DA, Krop I, Abramson V, Colleoni M, Traina T, Holmes F et al. Results from a randomized placebo-controlled phase 2 trial evaluating exemestane ± enzalutamide in patients with hormone receptor–positive breast cancer. Presented at the 2017 San Antonio Breast Cancer Symposium December 5–9, 2017: San Antonio,Texas USA [Google Scholar]
  • 88.Traina TA, Miller K, Yardley DA, O’Shaughnessy J, Cortes J, Awada A et al. (2015) Results from a phase 2 study of enzalutamide (ENZA), an androgen receptor (AR) inhibitor, in advanced AR+ triple-negative breast cancer (TNBC). J Clin Oncol 33(suppl); abstr 1003 [Google Scholar]
  • 89.Pfizer oncology to present data across 13 different types of cancer at ASCO 2017 annual meeting. [Accessed December 13, 2017] Available from: https://www.pfizer.com/news/press-release/press-release-detail/pfizer_oncology_to_present_data_across_13_different_types_of_cancer_at_asco_2017_annual_meeting
  • 90.Norris JD, Ellison SJ, Baker JG, Stagg DB, Wardell SE, Park S et al. (2017) Androgen receptor antagonism drives cytochrome P450 17A1 inhibitor efficacy in prostate cancer. J Clin Invest 127(6): 2326–2338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.FDA grants fast-track status for Innocrin’s seviteronel to treat metastatic CRPC. January 7, 2016. [Accessed March 13, 2017] Available from: http://www.pharmaceutical-technology.com/news/newsfda-grants-fast-track-status-innocrins-seviteronel-treat-metastatic-crpc-4770025
  • 92.Innocrin Pharmaceuticals appoints Fred Eshelman, PharmD as CEO and is granted Fast Track designation by FDA for seviteronel treatment of women with triple-negative breast cancer and women or men with estrogen receptor-positive breast cancer. [Accessed March 13, 2017] Available from: http://innocrinpharma.org/wp-content/uploads/2017/04/Innocrin-Pharmaceuticals-Appoints-Fred-Eshelman.pdf
  • 93.Gucalp A, Bardia A, Gabrail N, DaCosta N, Danso M, Elias AD et al. (2016) Phase 1/2 study of oral seviteronel (VT-464), a dual CYP17-lyase inhibitor and androgen receptor (AR) antagonist, in patients with advanced AR positive triple negative (TNBC) or estrogen receptor (ER) positive breast cancer (BC). Presented at the 2016 San Antonio Breast Cancer Symposium December 6–10, 2016: San Antonio, Texas USA Poster P2–08-04 [Google Scholar]
  • 94.Elias A, Gucalp A, Gardia A, Resaul A, Eisner J, Baskin-Bey E et al. (2017) Men with advanced breast cancer (BC): Initial phase (Ph) 2 clinical activity of seviteronel, a selective CYP17-lyase and androgen receptor inhibitor. Presented at the 2017 San Antonio Breast Cancer Symposium December 5–9, 2017: San Antonio, Texas USA Poster P5–23-04 [Google Scholar]

RESOURCES