a comparative biomarker study of 514 matched cases of male and female breast cancer reveals...

10
PRECLINICAL STUDY A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences Abeer M. Shaaban Graham R. Ball Rebecca A. Brannan Gabor Cserni Anna Di Benedetto Jo Dent Laura Fulford Helen Honarpisheh Lee Jordan J. Louise Jones Rani Kanthan Loaie Maraqa Maria Litwiniuk Marcella Mottolese Steven Pollock Elena Provenzano Philip R. Quinlan Georgina Reall Sami Shousha Mark Stephens Eldo T. Verghese Rosemary A. Walker Andrew M. Hanby Valerie Speirs Received: 3 October 2011 / Accepted: 25 October 2011 / Published online: 18 November 2011 Ó Springer Science+Business Media, LLC. 2011 Abstract Male breast cancer remains understudied despite evidence of rising incidence. Using a co-ordinated multi-centre approach, we present the first large scale biomarker study to define and compare hormone receptor profiles and survival between male and female invasive breast cancer. We defined and compared hormone receptor profiles and survival between 251 male and 263 female breast cancers matched for grade, age, and lymph node status. Tissue microarrays were immunostained for ERa, ERb1, -2, -5, PR, PRA, PRB and AR, augmented by HER2, CK5/6, 14, 18 and 19 to assist typing. Hierarchical clustering determined differential nature of influences between genders. Luminal A was the most common phe- notype in both sexes. Luminal B and HER2 were not seen in males. Basal phenotype was infrequent in both. No differences in overall survival at 5 or 10 years were observed between genders. Notably, AR-positive luminal A male breast cancer had improved overall survival over female breast cancer at 5 (P = 0.01, HR = 0.39, 95% CI = 0.26–0.87) but not 10 years (P = 0.29, HR = 0.75, 95% CI = 0.46–1.26) and both 5 (P = 0.04, HR = 0.37, 95% CI = 0.07–0.97) and 10 years (P = 0.04, HR = 0.43, 95% CI = 0.12–0.97) in the unselected group. Hierarchical clustering revealed common clusters between genders including total PR–PRA–PRB and ERb1/2 clus- ters. A striking feature was the occurrence of ERa on Presented in part at the 32nd Annual San Antonio Breast Cancer Symposium, 9-13 December, 2009, San Antonio, TX, Breast Cancer Research 2010, 18 May 2010, London UK and the Pathological Society of Great Britain and Ireland 2010 Summer Meeting, 30 June- 3 July, St Andrews, UK. A. M. Shaaban R. A. Brannan G. Reall E. T. Verghese A. M. Hanby St James’s Institute of Oncology, St James’s University Hospital, Leeds, UK G. R. Ball Nottingham Trent University, Nottingham, UK R. A. Brannan H. Honarpisheh L. Maraqa S. Pollock E. T. Verghese A. M. Hanby V. Speirs (&) Leeds Institute of Molecular Medicine, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK e-mail: [email protected] G. Cserni Bacs-Kiskun County Teaching Hospital, Nyiri ut 38, Kecskemet 6000, Hungary A. D. Benedetto M. Mottolese Regina Elena Cancer Institute, Rome, Italy J. Dent Calderdale Hospital, Halifax, UK L. Fulford Surrey & Sussex NHS Trust, Redhill, UK L. Jordan P. R. Quinlan University of Dundee/NHS Tayside, Dundee, UK J. L. Jones Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, London, UK R. Kanthan University of Saskatchewan, Saskatoon, Canada M. Litwiniuk Poznan University of Medical Sciences, Poznan, Poland 123 Breast Cancer Res Treat (2012) 133:949–958 DOI 10.1007/s10549-011-1856-9

Upload: et-al

Post on 26-Aug-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

PRECLINICAL STUDY

A comparative biomarker study of 514 matched cases of maleand female breast cancer reveals gender-specific biologicaldifferences

Abeer M. Shaaban • Graham R. Ball • Rebecca A. Brannan • Gabor Cserni •

Anna Di Benedetto • Jo Dent • Laura Fulford • Helen Honarpisheh •

Lee Jordan • J. Louise Jones • Rani Kanthan • Loaie Maraqa • Maria Litwiniuk •

Marcella Mottolese • Steven Pollock • Elena Provenzano • Philip R. Quinlan •

Georgina Reall • Sami Shousha • Mark Stephens • Eldo T. Verghese •

Rosemary A. Walker • Andrew M. Hanby • Valerie Speirs

Received: 3 October 2011 / Accepted: 25 October 2011 / Published online: 18 November 2011

� Springer Science+Business Media, LLC. 2011

Abstract Male breast cancer remains understudied

despite evidence of rising incidence. Using a co-ordinated

multi-centre approach, we present the first large scale

biomarker study to define and compare hormone receptor

profiles and survival between male and female invasive

breast cancer. We defined and compared hormone receptor

profiles and survival between 251 male and 263 female

breast cancers matched for grade, age, and lymph node

status. Tissue microarrays were immunostained for ERa,

ERb1, -2, -5, PR, PRA, PRB and AR, augmented by

HER2, CK5/6, 14, 18 and 19 to assist typing. Hierarchical

clustering determined differential nature of influences

between genders. Luminal A was the most common phe-

notype in both sexes. Luminal B and HER2 were not seen

in males. Basal phenotype was infrequent in both. No

differences in overall survival at 5 or 10 years were

observed between genders. Notably, AR-positive luminal

A male breast cancer had improved overall survival over

female breast cancer at 5 (P = 0.01, HR = 0.39, 95%

CI = 0.26–0.87) but not 10 years (P = 0.29, HR = 0.75,

95% CI = 0.46–1.26) and both 5 (P = 0.04, HR = 0.37,

95% CI = 0.07–0.97) and 10 years (P = 0.04, HR =

0.43, 95% CI = 0.12–0.97) in the unselected group.

Hierarchical clustering revealed common clusters between

genders including total PR–PRA–PRB and ERb1/2 clus-

ters. A striking feature was the occurrence of ERa on

Presented in part at the 32nd Annual San Antonio Breast Cancer

Symposium, 9-13 December, 2009, San Antonio, TX, Breast Cancer

Research 2010, 18 May 2010, London UK and the Pathological

Society of Great Britain and Ireland 2010 Summer Meeting, 30 June-

3 July, St Andrews, UK.

A. M. Shaaban � R. A. Brannan � G. Reall �E. T. Verghese � A. M. Hanby

St James’s Institute of Oncology, St James’s University Hospital,

Leeds, UK

G. R. Ball

Nottingham Trent University, Nottingham, UK

R. A. Brannan � H. Honarpisheh � L. Maraqa � S. Pollock �E. T. Verghese � A. M. Hanby � V. Speirs (&)

Leeds Institute of Molecular Medicine, Wellcome Trust Brenner

Building, University of Leeds, Leeds LS9 7TF, UK

e-mail: [email protected]

G. Cserni

Bacs-Kiskun County Teaching Hospital, Nyiri ut 38,

Kecskemet 6000, Hungary

A. D. Benedetto � M. Mottolese

Regina Elena Cancer Institute, Rome, Italy

J. Dent

Calderdale Hospital, Halifax, UK

L. Fulford

Surrey & Sussex NHS Trust, Redhill, UK

L. Jordan � P. R. Quinlan

University of Dundee/NHS Tayside, Dundee, UK

J. L. Jones

Barts Cancer Institute, Barts and The London School

of Medicine and Dentistry, London, UK

R. Kanthan

University of Saskatchewan, Saskatoon, Canada

M. Litwiniuk

Poznan University of Medical Sciences, Poznan, Poland

123

Breast Cancer Res Treat (2012) 133:949–958

DOI 10.1007/s10549-011-1856-9

Page 2: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

distinct clusters between genders. In female breast cancer,

ERa clustered with PR and its isoforms; in male breast

cancer, ERa clustered with ERb isoforms and AR. Our data

supports the hypothesis that breast cancer is biologically

different in males and females suggesting implications for

clinical management. With the incidence of male breast

cancer increasing this provides impetus for further study.

Keywords Male breast cancer � Hormone receptors �Androgen receptor � Hierarchical clustering

Introduction

According to figures from Cancer Research UK, there were

45,695 cases of female breast cancer (FBC) and 277 cases

of male breast cancer (MBC) diagnosed in the UK in 2007

[1]. In the US it was estimated that 1,970 men and 207,090

women would be diagnosed with breast cancer in 2010 [2].

Whilst MBC accounts for less than 1% of breast cancer

diagnoses worldwide, the overall improvements in survival

and mortality observed in FBC has not been seen to the

same extent in MBC, as demonstrated in a recent interro-

gation of the Surveillance Epidemiology and End Results

(SEER) database [3]. Moreover, the incidence rate of MBC

is rising steadily [4–7].

The etiology of MBC is poorly understood with most of

our current knowledge regarding its biology, natural his-

tory, and treatment extrapolated from FBC. Retrospective

studies are generally weakened by the small numbers of

cases available from any one centre with studies published

on as few as 15 cases [8], making it hard to draw biolog-

ically meaningful conclusions. It is, therefore, a challenge

to accrue sufficiently large numbers to allow comparative

analysis of possible prognostic or predictive biomarkers.

Many articles imply a general similarity of MBC to FBC

and this has resulted in MBC patients being treated in

exactly the same way as females in the clinic, which may

not be optimal. Survival rates for MBC are generally

assumed to be lower than FBC, probably as a result of later

diagnosis and the assumption that treatments which are

proven in FBC through clinical trials will have the same

impact in men [9].

A 40-year review of records of 759 cases from invasive

MBC from the US Armed Forces Institute of Pathology

database showed that the frequency of histological sub-

types in men was comparable to that of FBC, with the

exception of papillary carcinoma which was twice more

common in MBC [10]. To date, modern molecular sub-

typing has been reported in a single study of MBC where

only luminal A (35/42) and luminal B (7/42) subtypes were

observed [11].

Using a co-ordinated multi-centre approach, the aim of

this study was to conduct the first large scale study to

address and compare the expression profile of hormone

receptors and their effect on survival in FBC and MBC.

Methods

Patient cohorts

Following ethical approval from the Leeds (West) Research

Ethics Committee (06/Q1205/156), 514 formalin-fixed

paraffin-embedded blocks of male (251) and matched

female (263) breast cancers were obtained retrospectively.

The latter were all from Europe and the former from Europe

(n = 196) and Canada (n = 55). Informed consent was not

required as the anonymised material pre-dated September

2006, came from a Tissue Bank approved by the UK Human

Tissue Authority (or equivalent) or were from non-UK

patients. Patients had not received any therapy before sur-

gery. Details on adjuvant therapy were not extensively

available; where available this was predominantly endocrine

therapy (tamoxifen). Patient characteristics are presented in

Table 1. Cases were reviewed by specialised breast con-

sultant histopathologists (AMH, AMS, RAB) to confirm

histology and marked up for assembly into tissue micro-

arrays (TMAs) using 3 9 0.6 mm tissue cores per case

taken from formalin-fixed paraffin-embedded material as

previously described [10].

Immunohistochemistry (IHC)

Antibodies, dilution, and retrieval methods are listed in

Table 1. The antibody panel was focused on hormone

receptors oestrogen receptor (ER)a, ERb isoforms, pro-

gesterone receptor (PR) isoforms and androgen receptor

(AR) and additional biomarkers selected to distinguish

molecular subtypes of breast cancer (CK5/6, 14, 18,

HER2). Each marker was run as a batch with appropriate

positive (tissue known to express the biomarker of interest)

and negative (no primary antibody) controls. Scoring was

overseen by AMH, AMS, and RAB. Following visualisa-

tion of the signal with 3-30diaminobenzidine chromogen,

TMAs were digitised (Aperio Technologies), and hormone

E. Provenzano

Addenbrookes Hospital, Cambridge, UK

S. Shousha

Imperial College, London, UK

M. Stephens

University Hospital of North Staffordshire, Stoke-on Trent, UK

R. A. Walker

University of Leicester, Leicester, UK

950 Breast Cancer Res Treat (2012) 133:949–958

123

Page 3: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

receptor immunoreactivity was scored using the Allred

system with the following cut offs: ERa[ 2, ERb (and

isoforms) [ 3, AR [ 2, PR (and isoforms) [ 2, as vali-

dated in previous studies [12–14].

Hierachical clustering and principal components

analysis (PCA)

For hierarchical cluster analysis, IHC measurements were

used as inputs for all cases in each of the male and female

cohorts. A Euclidian distance measure was employed with

complete linkage of clusters. Clustering was conducted for

the data structure of cases with the immunohistochemical

parameters and for the immunohistochemical parameters as

they were expressed through the population. Cluster

dendrograms were plotted for both of these analyses for

each gender and compared. PCA was applied to the same

dataset. Analysis was based on covariances between

parameters and cases in the data. Variances were computed

based on the sum of squares/n - 1. Plots of the influences

of variables in the factor plane of the first and second and

the second and third principal components for both the

male and female cases were plotted separately. As with the

hierarchical clustering, both the male and female cases

were combined into a single data set. The influence of

variables in the factor plane of the combined cases were

plotted for the first and second and the second and third

principal components. The distributions of cases within the

combined sets were also plotted.

Statistical analysis

Patient and disease characteristics were compared between

male and FBCs using the v2 test (GraphPad). Associations

with disease-free and overall survival (DFS and OS,

respectively) were analysed by Kaplan–Meier plots and log

rank test. P-values were two-sided, and P \ 0.05 was

considered significant.

Results

A total of 514 cases of breast carcinoma were studied,

including 251 males and 263 females. The median age for

the male cohort was 66 years (range 30–94) and 59 years

(range 27–92) for females. Patient characteristics are

shown in Table 2. As this was a matched cohort, no sig-

nificant differences were observed in grade, or lymph node

status between genders. Significant differences were

observed in the distribution of histopathological subtypes

(P \ 0.0001). There was an even distribution of ductal

phenotype whilst lobular carcinomas found in 9% of the

female cohort was only seen in a single male case. Papil-

lary and mucinous phenotypes were restricted to males. A

significantly higher proportion of males expressed ERacompared to females (80 and 68%, respectively), although

no differences in the frequency of PR was observed (71 and

72%, respectively). Follow up data was available on 183

(73%) male and 237 (90%) female cases.

Both cohorts were classified into molecular subtypes by

IHC: luminal A (ERa?, and/or PR?, HER2-), luminal B

(ERa?, and/or PR?, HER2?), HER2 (ERa, PR-, HER2?)

and basal-like (ERa-, PR-, HER2-, CK5/6?) according to

previous studies [15–17]. Representative immunopro-

files for each subgroup are shown in Fig. 1. Significant

Table 1 Patient characteristics of male and female breast cancers

Characteristic Male

(n = 251)

no (%)

Female

(n = 263)

no (%)

P values

Histology \0.0001

Ductal 208 (83) 220 (84)

Lobular 1 (0.4) 23 (9)

Papillary 11 (4) 0

Intraductal papillary 4 (1.5) 0

Micropapillary 1 (0.4) 0

Mucinous 9 (4) 0

Mixed 13 (5) 14 (7)

Unknown 4 (1.6) 0

Grade 0.948

1 25 (10) 29 (11)

2 128 (51) 140 (53)

3 81 (33) 94 (36)

Unknown 17 (6) 0

Lymph node 0.521

? 76 (30) 131 (50)

- 80 (32) 121 (46)

Unknown 95 (48) 11 (4)

ERa \0.0001

? 201 (80) 180 (68)

- 22 (9) 56 (21)

Unknown 28 (11) 27 (10)

PR 0.838

? 177 (71) 190 (72)

- 39 (15) 44 (17)

Unknown 35 (14) 29 (11)

Subtypea 0.0004

Luminal A 199 (98) 197 (90)

Luminal B 0 (0) 14 (6)

Basal 4 (2) 4 (2)

HER2?/ERa- 0 5 (2)

a Subtype classification was not possible in cases from 48 males and

43 females due to core loss from the section for at least one of the

biomarkers, or less commonly, exhaustion of the TMA core

Breast Cancer Res Treat (2012) 133:949–958 951

123

Page 4: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

differences were observed between molecular subtypes

(P = 0.0004). Luminal A was seen in 98% of males and 90%

of females. Luminal B or HER2 subgroups were not

observed in males but found in 6 and 2% of females,

respectively. Basal-like tumours (ERa-, PR-, HER2-,

CK5/6?) were infrequent in both cohorts (2% in each).

We then examined the frequencies of expression

between genders of other hormone receptors including AR,

nuclear and cytoplasmic ERb1 and ERb2, nuclear ERb5,

plus the PR isoforms A and B (Table 3). AR immunore-

activity was expressed in 64% of males and 93% females,

respectively, (P \ 0.0001). For ERb1 and -2, both nuclear

and cytoplasmic immunoreactivity was assessed [12].

ERb1 nuclear immunoreactivity was significantly expres-

sed in FBC whilst cytoplasmic ERb1 and ERb2 immuno-

reactivity were associated with MBC. No associations were

observed for ERb5. Of the PR isoforms, only PRA was

significantly expressed in MBC. As the male cohort con-

tained cases of European and Canadian origin, we tested if

there were differences between these; none were found.

In luminal A carcinomas, no differences in overall sur-

vival were observed between genders at either 5 or

10 years (Fig. 2a, b). This was also reflected in the unse-

lected cohorts (data not shown). When hormone receptor

expression was considered, only AR significantly associ-

ated with survival. AR-positive luminal A MBC had sig-

nificantly improved overall survival over the equivalent

FBC at 5 (P = 0.01, HR = 0.39, 95% CI = 0.26–0.87) but

not 10 (P = 0.29, HR = 0.75, 95% CI = 0.46–1.26) years

(Fig. 2c, d). In the unselected group, ERa and AR-positive

MBC had significantly improved overall survival over ERaand AR-negative cases at both 5 (P = 0.04, HR = 0.37,

95% CI = 0.07–0.97) and 10 (P = 0.04, HR = 0.43, 95%

CI = 0.12–0.97) years (Fig. 2e, f) with ERa and AR-

positive MBC also having significantly improved overall

survival over the equivalent FBCs at 5 (P = 0.05,

HR = 0.48, 95% CI = 0.29–1.00) but not 10 (P = 0.37,

HR = 0.79, 95% CI = 0.48–1.32) years (Fig. 2g, h).

Hierarchical clustering based on hormone receptor profiles

classified MBC and FBC into three distinct groups (Fig. 3).

The cytoplasmic ERb cluster was common to both genders. In

FBC an ERa/PR cluster was observed, grouping ERa and PR

isoforms, whilst ERb isoforms clustered with AR (ERb/AR

cluster). In MBC, there were striking changes in the position

of ERa; AR and ERa clustered with ERb (ERa/b AR cluster)

whilst PR isoforms formed an independent cluster (PR clus-

ter). This was also reflected in a PCA-based plot of variable

factor co-ordinates (data not shown).

Discussion

Currently, MBC is treated based on the assumption that it

is essentially the same disease as FBC. In this the largest

comparative study to date, directly comparing the immu-

nohistochemical profile of matched MBCs and FBCs has

revealed that whilst superficially there is similarity between

genders, when probed more deeply, subtle differences are

uncovered.

The histological breakdown of our cohort is in line with

previous reports with the papillary phenotype and variants

thereof, which are twice more common in males, only seen

in the male cohort [10]. Although we did not observe any

mucinous carcinomas in our female cohort the expected

frequency of this phenotype is only 0.9% [18]. We observed

only a single case of lobular carcinoma in males; this is to be

expected given the rarity of this phenotype in men [19].

Thus, our cohort can be regarded as representative.

Table 2 Details of antibodies used for immunohistochemical analysis

Antibody Clone Supplier Dilution Secondary detection Pre-treatment

ERa 1D5 DAKO 1:100 DAKO EnVision High pressure heat retrieval

in Vector low pH antigen

unmasking solutionERb1 PPG5/10 Serotec 1:20

ERb2 57/3 Serotec 1:20

ERb5 5/25 Serotec 1:50

Total PR PgR636 DAKO 1:200

PRA 16 Novacastra RTU

PRB San27 Novacastra RTU

AR AR441 DAKO 1:200

CK5/6 D5/16 B4 DAKO 1:100

CK14 LL002 Serotec 1:50

CK18 CY-90 Sigma 1:500

CK19 RCK108 DAKO 1:150

RTU ready to use

952 Breast Cancer Res Treat (2012) 133:949–958

123

Page 5: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

We used IHC to classify our breast tumours into

molecular subtypes. Whilst gene array analysis is still

considered the ‘gold standard’, we were unable to apply

this to our 514-case cohort due to high cost. Nevertheless,

molecular profiling of breast cancer based on immunohis-

tochemical typing has now gained widespread acceptance

as a surrogate method and is arguably more robust as it

overcomes the limitations of gene array in that it directly

identifies the cells expressing the marker of interest [15–17,

20]. When applied to TMAs, a limitation is core loss,

which we experienced in this study and which may have

impacted on the higher than anticipated levels of luminal A

phenotype in the female population. Nevertheless, luminal

A phenotype was the most common in both sexes with

basal-like tumours infrequent in both. Sporadic expression

of basal cytokeratins has been previously reported in MBC

[21]. Interestingly, the luminal B phenotype was not seen

in males. This contrasts a recent study of 42 MBCs where

luminal B was seen in 17% of cases [11]. The lack of

luminal B carcinomas in our male cohort reflects the

absence of HER2 expression, which has been variably

reported in MBC ranging from 0 to 95% [22]. It is notable

that some of the earlier studies on HER2 relied solely on

IHC to determine positivity, considering any degree of

membrane immunoreactivity positive [8, 22–28]. The

validity of studies relying on HER2 IHC without recogni-

tion of gene expression is questionable. According to

ASCO/ACP and NEQAS guidelines only those scoring 3?

or above are considered HER2 positive. Equivocal cases

are scored 2? and go forward for FISH analysis and only

those with HER2 gene amplification are considered posi-

tive. In studies using both IHC and FISH to detect HER2,

protein expression was always higher than gene amplifi-

cation [29–31]. Although we relied on IHC to detect

HER2, we are confident our data is robust; we are a

regional HER2 testing centre and our HER2 IHC was

conducted via this service using two different antibodies

and running the test according to clinical standards. Whilst

ERα PR HER2 CK5/6

Not

applicable

Not

applicable

Not

applicable

a

b

c

d

Fig. 1 Semi-serial sections from male (a, c) and female (b, d) breast

carcinoma TMAs showing immunoprofiles for each molecular

subgroup. a luminal A (ERa?, PR?, HER2-), b luminal B

(ERa?, PR?, HER2?), c basal (ERa-, PR-, HER2-, CK5/6?),

d HER2 (ERa-, PR-, HER2?). Original magnification = 910

(TMA core) and 940 (insets)

Breast Cancer Res Treat (2012) 133:949–958 953

123

Page 6: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

we did not observe any cases which scored [2? in our

MBC cohort, we observed scores of 1? in 22/251 cases

(8%). These would be considered negative in clinical

practice. Nevertheless, this provides confidence that the

high frequency of HER2 negativity we observed was not

simply due to antigen degradation in archival material.

It has been suggested that separation of luminal A and

luminal B breast tumours should be based on the expression

of proliferation markers such as Ki67, not on HER2

expression as is currently the case [32]. However, this has

yet to gain widespread acceptance and we believe there are

several issues that still need to be standardised before this

can be implemented. These include choice of Ki67 antibody,

e.g. MIB1 or SP6 [33] and how to optimally distinguish

between low and high proliferation scores. Once these issues

are resolved it will be interesting to determine if the differ-

ence between MBC and FBC in this series is purely the result

of lack of HER2 expression, or if MBCs have a lower pro-

liferative index as well. Contrary to the general impression,

one of the most significant findings from this, the most

authoritative study to date, was the observation of no dif-

ferences in overall survival at 5 or 10 years between genders

in either our unselected or the luminal A cohorts. Whilst

there have been a number of case–control and population-

based studies addressing survival in MBC using data from

cancer registries [3, 4, 34, 35], direct comparative studies

between genders are scarce. A Chinese study of 35 MBC and

70 matched FBC showed the latter had significantly better

overall survival at 5 and 10 years, but when the comparison

was restricted to female postmenopausal, outcomes were

similar [36]. Despite a small number of cases in the male arm

and unbalanced cohort size, a Japanese study of 14 MBC and

140 FBC showed no difference in overall survival [37]. This

was also reflected in a UK study comparing outcome in 41

MBC and 123 FBC which showed that when matched for

key prognostic factors (size, grade and lymph node status),

outcome was similar between genders [38], agreeing with

our study. Of note was the observation that when MBC was

compared with an unselected FBC group, males had worse

outcome [38], which may explain some of the earlier studies

inferring a worse prognosis in men [39, 40]. Gender com-

parative information obtained from 1988 to 2003 SEER data

showed worse breast cancer-specific survival in males

diagnosed with stage I disease; however, the authors

attributed this to in-stage migration rather than being of

clinical relevance [41].

A common finding in MBC is the higher frequency of

hormone receptor expression in particular ERa (reviewed

in [42]), which was also reflected in this study. We have

explored this further using hierarchical clustering where

one of the striking features was the occurrence of ERa on

distinct clusters in males and females. In FBC, ERa clus-

tered with PR and its isoforms; as PR is oestrogen-regu-

lated [43], this is unsurprising. In MBC, the position of

ERa changed, clustering instead with ERb isoforms and

AR. The potential role of ERb in breast cancer has been the

subject of much debate and recent work in FBC by us and

others shows this depends on the cell location and the type

of isoforms expressed [9, 44]. However, ERb is subject to

complex regulation involving 30UTRs [45] and microRNAs

[46], and we need to further understand its biology before

speculating on any role it may play in MBC.

Regarding the potential relationship of AR with ERa, at

a functional level, AR transfection into ERa-positive breast

cancer cells inhibited ERa transactivation and oestrogen-

stimulated growth through interaction with oestrogen

response elements [47]. Other work indicates that oestro-

gen activation via ERa and ERb can mediate AR signalling

[48]. Given the recognised pro-proliferative effects of ERaand PR and the anti-proliferative effects of ERb and AR

[49] this suggests the coordinated expression of these

receptors could influence survival. This was demonstrated

Table 3 Expression of AR, ERb, and PR isoforms in male and

female luminal A breast carcinoma

Biomarker Negative

(%)

Positive

(%)

P value OR (95% CI)

AR

Male 86 113 \0.0001 0.12 (0.06–0.208)

Female 16 181

PRA

Male 47 152 \0.0001 3.47 (2.25–5.34)

Female 102 95

PRB

Male 79 120 0.486 1.18 (0.80–1.76)

Female 86 111

ERb1 (nuc)

Male 77 122 \0.0001 0.14 (0.08–0.25)

Female 16 181

ERb1 (cyto)a

Male 26 157 \0.0001 22.34 (12.99–38.43)

Female 148 40

ERb2 (nuc)

Male 38 161 0.36 1.29 (0.79–2.09)

Female 46 151

ERb2 (cyto)b

Male 63 117 \0.0001 2.50 (1.64–3.80)

Female 109 81

ERb5 (nuc)

Male 49 150 0.20 0.71 (0.44–1.15)

Female 37 160

Nuc nuclear expression, cyto cytoplasmic expressiona 16 male and 9 female and b 19 male and 7 female cases were

unscoreable due to core loss/exhaustion

954 Breast Cancer Res Treat (2012) 133:949–958

123

Page 7: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

0 10 20 30 40 50 600

25

50

75

100

P = 0.09

MonthsP

erce

nt

surv

ival

0 20 40 60 80 100 1200

25

50

75

100

P = 0.42

Months

Per

cen

t su

rviv

al

0 10 20 30 40 50 600

25

50

75

100

P = 0.01

Months

Per

cen

t su

rviv

al

0 20 40 60 80 100 1200

25

50

75

100

P = 0.29

Months

Per

cen

t su

rviv

al

0 10 20 30 40 50 600

25

50

75

100

P = 0.04

Months

Per

cen

t su

rviv

al

0 20 40 60 80 100 1200

25

50

75

100

P = 0.04

Months

Per

cen

t su

rviv

al

0 10 20 30 40 50 600

25

50

75

100

P = 0.05

Months

Per

cen

t su

rviv

al

0 20 40 60 80 100 1200

25

50

75

100

P = 0.37

Months

Per

cen

t su

rviv

al

a b

c

e

g

d

f

h

15/119

47/185

25/119

69/177

7/79

46/178

16/77

67/170

7/75

44/182

17/80

69/193

5/57

6/27

17/80

8/22

Fig. 2 Kaplan–Meier survival

curves comparing survival

between genders according to

different tumour classifications.

No gender-related differences

were observed comparing OS of

MBC (blue line) and FBC (pinkline) at 5 (a) and 10 (b) years.

Comparison of OS of Luminal

A AR-positive MBC (blue line)

and FBC (pink line) showed

MBC had significantly

improved OS at 5 (c) but not 10

(d) years. In unselected ERa-

positive MBC those which were

AR positive (black line) had

significantly improved over

those who were AR negative

(orange line) at both 5 (e) and

10 (f) years. Comparison of OS

of unselected ERa and AR-

positive carcinomas showed

MBC (blue line) had

significantly improved survival

over FBC (pink line) at 5 (g) but

not 10 (h) years

Breast Cancer Res Treat (2012) 133:949–958 955

123

Page 8: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

in the current study where AR-positive luminal A MBC

had improved overall survival than the equivalent FBC and

was also borne out in the unselected group. This supports

other recent work showing AR is an important prognostic

factor in ERa-positive FBC [12]. Previous studies exam-

ining the impact of AR in MBC have been compounded by

the high expression frequency of AR in some studies [22],

the low number of cases in others [50], or combinations of

both [51, 52] precluding meaningful survival analysis.

Studies examining the effect of AR on survival in MBC are

contradictory, possibly as a result of limited numbers

available from single centre studies. In a series of 43 MBC

Kwiatowska et al. [53] showed AR expression correlated

with reduced survival. A similar-sized study (n = 47)

showed no effects of AR expression in MBC on survival,

however, it is noteworthy that in the study, MIB-1

expression, which detects proliferating cells, was higher in

AR negative compared to AR-positive cases [26]. The

association of AR positivity with better outcome in MBC

in this study indicates this is a potentially important

prognostic factor, paralleling observations in FBC where

the potential prognostic role for AR in FBC is receiving

increased attention [54] following the observation that AR

is an independent prognostic marker in a large series of

FBCs (n = 953; [12]). Moreover, AR expression in MBC

could turn out to have both prognostic and predictive value;

its presence suggests that anti-androgen therapy could be

explored as a therapeutic approach. Androgen blockade is

commonly used in prostate cancer treatment but so far

remains inadequately tested in breast cancer [55]. The

complete absence of cell line models derived from MBCs

presents a challenge in being able to model this in vitro.

To our knowledge, this is the largest retrospective bio-

marker study directly comparing matched male and female

breast carcinoma; however, we acknowledge there are

some limitations. Despite our best efforts, these include

lack of availability of follow up data and missing/

incomplete clinicopathological data. Of note is the absence

of data on germline mutations, particularly BRCA2 which

is involved in MBC development and associated with

reduced survival [53, 56, 57]. Nevertheless, our study has

confirmed that whilst superficially similar to FBC, when

studied more rigorously MBC is biologically different,

echoing a hypothesis proposed by the Multidisciplinary

Meeting on MBC [58] and supporting recent work at the

transcriptional [59], microRNA [60, 61] and genomic [62]

levels. With the incidence of MBC rising [4–7], collec-

tively these studies provide a strong impetus for further

study of this rare cancer type.

Acknowledgments Thanks to the Tayside Tissue Bank for kindly

providing some of the MBC cases. This study was supported by the

Breast Cancer Campaign (UK Charity no. 05074725).

Conflict of interest None.

References

1. Breast cancer-survival statistics. Cancer Research UK Web Site.

http://info.cancerresearchuk.org/cancerstats/. Accessed 26 Jan

2011

2. Jemal A, Siegel R, Xu J, Ward E (2010) Cancer statistics. CA

Cancer J Clin 60:277–300

3. Anderson WF, Jatoi I, Tse J, Rosenberg PS (2010) Male breast

cancer: a population-based comparison with female breast cancer.

J Clin Oncol 28:232–239

4. Giordano SH, Cohen DS, Buzdar AU, Perkins G, Hortobagyi GN

(2004) Breast carcinoma in men: a population-based study.

Cancer 101:51–57

5. Hodgson NC, Button JH, Franceschi D, Moffat FL, Livingstone

AS (2004) Male breast cancer: is the incidence increasing? Ann

Surg Oncol 11:751–755

6. Speirs V, Shaaban AM (2009) The rising incidence of male breast

cancer. Breast Cancer Res Treat 115:429–430

7. White J, Kearins O, Dodwell D, Horgan K, Hanby AM, Speirs V

(2011) Male breast carcinoma: increased awareness needed.

Breast Cancer Res 13:219

Fig. 3 Hierarchical cluster analysis showing relatedness of hormone receptor profiles between genders. Three clusters were observed in each

gender (dotted lines). The cERb cluster was common to both but there was a shift in the position of ERa in MBC

956 Breast Cancer Res Treat (2012) 133:949–958

123

Page 9: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

8. Clark JL, Nguyen PL, Jaszcz WB, Jatoi A, Niehans GA (2000)

Prognostic variables in male breast cancer. Am Surg 66:502–511

9. Fentiman IS, Fourquet A, Hortobagyi GN (2006) Male breast

cancer. Lancet 367:595–604

10. Burga AM, Fadare O, Lininger RA, Tavassoli FA (2006) Invasive

carcinomas of the male breast: a morphologic study of the dis-

tribution of histologic subtypes and metastatic patterns in 778

cases. Virchows Arch 449:507–512

11. Ge Y, Sneige N, Eltorky MA, Wang Z, Lin E, Gong Y, Guo M

(2009) Immunohistochemical characterization of subtypes of

male breast carcinoma. Breast Cancer Res 11:R28

12. Shaaban AM, Green AR, Karthik S, Alizadeh Y, Hughes TA,

Harkins L, Ellis IO, Robertson JF, Paish EC, Saunders PT,

Groome NP, Speirs V (2008) Nuclear and cytoplasmic expression

of ERb1, ERb2, and ERb5 identifies distinct prognostic outcome

for breast cancer patients. Clin Cancer Res 14:5228–5235

13. Leake R, Barnes D, Pinder S, Ellis I, Anderson L, Anderson T,

Adamson R, Rhodes T, Miller K, Walker R (2000) Immunohis-

tochemical detection of steroid receptors in breast cancer: a

working protocol. UK Receptor Group, UK NEQAS, The Scot-

tish Breast Cancer Pathology Group, and The Receptor and

Biomarker Study Group of the EORTC. J Clin Pathol 53:634–635

14. Castellano I, Allia E, Accortanzo V, Vandone AM, Chiusa L,

Arisio R, Durando A, Donadio M, Bussolati G, Coates AS, Viale

G, Sapino A (2010) Androgen receptor expression is a significant

prognostic factor in estrogen receptor positive breast cancers.

Breast Cancer Res Treat 124:607–617

15. Callagy G, Cattaneo E, Daigo Y, Happerfield L, Bobrow LG,

Pharoah PD, Caldas C (2003) Molecular classification of breast

carcinomas using tissue microarrays. Diagn Mol Pathol 12:27–34

16. Abd El-Rehim DM, Ball G, Pinder SE, Rakha E, Paish C, Rob-

ertson JF, Macmillan D, Blamey RW, Ellis IO (2005) High-

throughput protein expression analysis using tissue microarray

technology of a large well-characterised series identifies biolog-

ically distinct classes of breast cancer confirming recent cDNA

expression analyses. Int J Cancer 116:340–350

17. Blows FM, Driver KE, Schmidt MK, Broeks A, van Leeuwen FE,

Wesseling J, Cheang MC, Gelmon K, Nielsen TO, Blomqvist C,

Heikkila P, Heikkinen T, Nevanlinna H, Akslen LA, Begin LR,

Foulkes WD, Couch FJ, Wang X, Cafourek V, Olson JE, Ba-

glietto L, Giles GG, Severi G, McLean CA, Southey MC, Rakha

E, Green AR, Ellis IO, Sherman ME, Lissowska J, Anderson WF,

Cox A, Cross SS, Reed MW, Provenzano E, Dawson SJ, Dunning

AM, Humphreys M, Easton DF, Garcıa-Closas M, Caldas C,

Pharoah PD, Huntsman D (2010) Subtyping of breast cancer by

immunohistochemistry to investigate a relationship between

subtype and short and long term survival: a collaborative analysis

of data for 10,159 cases from 12 Studies. PLoS Med 7:e1000279

18. Ellis IO, Galea M, Broughton N, Locker A, Blamey RW, Elston

CW (1992) Pathological prognostic factors in breast cancer. II.

Histological type. Relationship with survival in a large study with

long-term follow-up. Histopathology 20:479–489

19. Michaels BM, Nunn CR, Roses DF (1994) Lobular carcinoma of

the male breast. Surgery 115:402–405

20. O’Brien KM, Cole SR, Tse CK, Perou CM, Carey LA, Foulkes

WD, Dressler LG, Geradts J, Millikan RC (2010) Intrinsic breast

tumor subtypes, race, and long-term survival in the Carolina

Breast Cancer Study. Clin Cancer Res 16:6100–6110

21. Ciocca V, Bombonati A, Gatalica Z, Di Pasquale M, Milos A,

Ruiz-Orrico A, Dreher D, Folch N, Monzon F, Santeusanio G,

Perou CM, Bernard PS, Palazzo JP (2006) Cytokeratin profiles of

male breast cancers. Histopathology 49:365–370

22. Rayson D, Erlichman C, Suman VJ, Roche PC, Wold LE, Ingle

JN, Donohue JH (1998) Molecular markers in male breast car-

cinoma. Cancer 83:1947–1955

23. Blin N, Kardas I, Welter C, Rys J, Niezabitowski A, Limon J,

Seitz G (1993) Expression of the c-erbB2 proto-oncogene in male

breast carcinoma: lack of prognostic significance. Oncology

50:401–408

24. Avisar E, McParland E, Dicostanzo D, Axelrod D (2006) Prog-

nostic factors in node-negative male breast cancer. Clin Breast

Cancer 7:331–335

25. Shpitz B, Bomstein Y, Sternberg A, Klein E, Liverant S, Grois-

man G, Bernheim J (2000) Angiogenesis, p53, and c-erbB-2

immunoreactivity and clinicopathological features in male breast

cancer. J Surg Oncol 75:252–257

26. Pich A, Margaria E, Chiusa L, Candelaresi G, Dal Canton O

(1999) Androgen receptor expression in male breast carcinoma:

lack of clinicopathological association. Br J Cancer 79:959–964

27. Bruce DM, Heys SD, Payne S, Miller ID, Eremin O (1996) Male

breast cancer: clinico-pathological features, immunocytochemi-

cal characteristics and prognosis. Eur J Surg Oncol 22:42–46

28. Dawson PJ, Paine TM, Wolman SR (1992) Immunocytochemical

characterization of male breast cancer. Mod Pathol 5:621–625

29. Bloom KJ, Govil H, Gattuso P, Reddy V, Francescatti D (2001)

Status of HER-2 in male and female breast carcinoma. Am J Surg

182:389–392

30. Fonseca RR, Tomas AR, Andre S, Soares J (2006) Evaluation of

ERBB2 gene status and chromosome 17 anomalies in male breast

cancer. Am J Surg Pathol 30:1292–1298

31. Rudlowski C, Friedrichs N, Faridi A, Fuzesi L, Moll R, Bastert G,

Rath W, Buttner R (2004) Her-2/neu gene amplification and

protein expression in primary male breast cancer. Breast Cancer

Res Treat 84:215–223

32. Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J,

Watson M, Davies S, Bernard PS, Parker JS, Perou CM, Ellis MJ,

Nielsen TO (2009) Ki67 index, HER2 status, and prognosis of

patients with luminal B breast cancer. J Natl Cancer Inst

101:736–750

33. Zabaglo L, Salter J, Anderson H, Quinn E, Hills M, Detre S,

A’Hern R, Dowsett M (2010) Comparative validation of the SP6

antibody to Ki67 in breast cancer. J Clin Pathol 63:800–804

34. Anderson WF, Althuis MD, Brinton LA, Devesa SS (2004) Is

male breast cancer similar or different than female breast cancer?

Breast Cancer Res Treat 83:77–86

35. Adami HO, Hakulinen T, Ewertz M, Tretli S, Holmberg L,

Karjalainen S (1989) The survival pattern in male breast cancer.

An analysis of 1429 patients from the Nordic countries. Cancer

64:1177–1182

36. Xia LP, Zhou FF, Guo GF, Wang F, Wang X, Yuan ZY, Zhang B

(2010) Chinese female breast cancer patients show a better

overall survival than their male counterparts. Chin Med J (Engl)

123:2347–2352

37. Anan K, Mitsuyama S, Nishihara K, Abe Y, Iwashita T, Ihara T,

Tamae K, Ono M, Toyoshima S (2004) Breast cancer in Japanese

men: does sex affect prognosis? Breast Cancer 11:180–186

38. Willsher PC, Leach IH, Ellis IO, Bourke JB, Blamey RW,

Robertson JF (1997) A comparison outcome of male breast

cancer with female breast cancer. Am J Surg 173:185–188

39. Ouriel K, Lotze MT, Hinshaw JR (1984) Prognostic factors of

carcinoma of the male breast. Surg Gynecol Obstet 159:373–376

40. Spence RA, MacKenzie G, Anderson JR, Lyons AR, Bell M

(1985) Long-term survival following cancer of the male breast in

Northern Ireland. A report of 81 cases. Cancer 55:648–652

41. Gnerlich JL, Deshpande AD, Jeffe DB, Selam S, Kimbuende E,

Margenthaler JA (2010) Poorer survival outcomes for male breast

cancer compared with female breast cancer may be attributable to

in-stage migration. Ann Surg Oncol 18:1837–1844

42. Nahleh Z, Girnius S (2006) Male breast cancer: a gender issue.

Nat Clin Pract Oncol 3:428–437

Breast Cancer Res Treat (2012) 133:949–958 957

123

Page 10: A comparative biomarker study of 514 matched cases of male and female breast cancer reveals gender-specific biological differences

43. Weigel MT, Dowsett M (2010) Current and emerging biomarkers

in breast cancer: prognosis and prediction. Endocr Relat Cancer

17:R245–R262

44. Honma N, Horii R, Iwase T, Saji S, Younes M, Takubo K,

Matsuura M, Ito Y, Akiyama F, Sakamoto G (2008) Clinical

importance of estrogen receptor-b evaluation in breast cancer

patients treated with adjuvant tamoxifen therapy. J Clin Oncol

26:3727–3734

45. Smith L, Coleman LJ, Cummings M, Satheesha S, Shaw SO,

Speirs V, Hughes TA (2010) Expression of oestrogen receptor bisoforms is regulated by transcriptional and post-transcriptional

mechanisms. Biochem J 429:283–290

46. Al-Nakhle H, Burns PA, Cummings M, Hanby AM, Hughes TA,

Satheesha S, Shaaban AM, Smith L, Speirs V (2010) Estrogen

receptor b1 expression is regulated by miR-92 in breast cancer.

Cancer Res 70:4778–4784

47. Peters AA, Buchanan G, Ricciardelli C, Bianco-Miotto T, Cen-

tenera MM, Harris JM, Jindal S, Segara D, Jia L, Moore NL,

Henshall SM, Birrell SN, Coetzee GA, Sutherland RL, Butler

LM, Tilley WD (2009) Androgen receptor inhibits estrogen

receptor-alpha activity and is prognostic in breast cancer. Cancer

Res 69:6131–6140

48. Narita D, Anghel A, Cimpean AM, Izvernariu D, Cireap N, Ilina R,

Ursoniu S (2010) Interaction between estrogens and androgen

receptor genes microsatellites, prostate-specific antigen and andro-

gen receptor expressions in breast cancer. Neoplasma 57:198–206

49. Conzen SD (2008) Minireview: nuclear receptors and breast

cancer. Mol Endocrinol 22:2215–2228

50. Munoz de Toro MM, Maffini MV, Kass L, Kass L, Luque EH

(1998) Proliferative activity and steroid hormone receptor status

in male breast carcinoma. J Steroid Biochem Mol Biol 67:

333–339

51. Murphy CE, Carder PJ, Lansdown MR, Speirs V (2006) Steroid

hormone receptor expression in male breast cancer. Eur J Surg

Oncol 32:44–47

52. Sasano H, Kimura M, Shizawa S, Kimura N, Nagura H (1996)

Aromatase and steroid receptors in gynecomastia and male breast

carcinoma: an immunohistochemical study. J Clin Endocrinol

Metab 81:3063–3067

53. Kwiatkowska E, Teresiak M, Filas V, Karczewska A, Bre-

borowicz D, Mackiewicz A (2003) BRCA2 mutations and

androgen receptor expression as independent predictors of out-

come of male breast cancer patients. Clin Cancer Res 9:

4452–4459

54. Higgins MJ, Wolff AC (2010) The androgen receptor in breast

cancer: learning from the past. Breast Cancer Res Treat 124:

619–621

55. Folkerd EJ, Dowsett M (2010) Influence of sex hormones on

cancer progression. J Clin Oncol 28:4038–4044

56. Thorlacius S, Olafsdottir G, Tryggvadottir L, Neuhausen S,

Jonasson JG, Tavtigian SV, Tulinius H, Ogmundsdottir HM,

Eyfjord JE (1996) A single BRCA2 mutation in male and female

breast cancer families from Iceland with varied cancer pheno-

types. Nat Genet 13:117–119

57. Ding YC, Steele L, Kuan CJ, Greilac S, Neuhausen SL (2011)

Mutations in BRCA2 and PALB2 in male breast cancer cases

from the United States. Breast Cancer Res Treat 126:771–778

58. Korde LA, Zujewski JA, Kamin L, Giordano S, Domchek S,

Anderson WF, Bartlett JM, Gelmon K, Nahleh Z, Bergh J, Cutuli B,

Pruneri G, McCaskill-Stevens W, Gralow J, Hortobagyi G, Cardoso

F (2010) Multidisciplinary meeting on male breast cancer: summary

and research recommendations. J Clin Oncol 28:2114–2122

59. Callari M, Cappelletti V, De Cecco L, Musella V, Miodini P,

Veneroni S, Gariboldi M, Pierotti MA, Daidone MG (2011) Gene

expression analysis reveals a different transcriptomic landscape

in female and male breast cancer. Breast Cancer Res Treat

127:601–610

60. Fassan M, Baffa R, Palazzo JP, Lloyd J, Crosariol M, Liu CG,

Volinia S, Alder H, Rugge M, Croce CM, Rosenberg A (2009)

MicroRNA expression profiling of male breast cancer. Breast

Cancer Res 11:R58

61. Lehmann U, Streichert T, Otto B, Albat C, Hasemeier B,

Christgen H, Schipper E, Hille U, Kreipe HH, Langer F (2010)

Identification of differentially expressed microRNAs in human

male breast cancer. BMC Cancer 10:109

62. Orr N, Cooke R, Jones M, Fletcher O, Dudbridge F, Chilcott-

Burns S, Tomczyk K, Broderick P, Houlston R, Ashworth A,

Swerdlow A (2011) Genetic variants at chromosomes 2q35,

5p12, 6q25.1, 10q26.13, and 16q12.1 influence the risk of breast

cancer in men. Plos Genet 7:e1002290

958 Breast Cancer Res Treat (2012) 133:949–958

123