Skip to main content
Erschienen in: Breast Cancer Research and Treatment 1/2013

Open Access 01.07.2013 | Brief Report

Clinical–pathologic significance of cancer stem cell marker expression in familial breast cancers

verfasst von: Anita Bane, Alicia Viloria-Petit, Dushanthi Pinnaduwage, Anna Marie Mulligan, Frances P. O’Malley, Irene L. Andrulis

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 1/2013

Abstract

Human breast cancer cells with a CD44+/CD24−/low or ALDH1+ phenotype have been demonstrated to be enriched for cancer stem cells (CSCs) using in vitro and in vivo techniques. The aim of this study was to determine the association between CD44+/CD24−/low and ALDH1 expression with clinical–pathologic tumor characteristics, tumor molecular subtype, and survival in a well characterized collection of familial breast cancer cases. 364 familial breast cancers from the Ontario Familial Breast Cancer Registry (58 BRCA1-associated, 64 BRCA2-associated, and 242 familial non-BRCA1/2 cancers) were studied. Each tumor had a centralized pathology review performed. TMA sections of all tumors were analyzed for the expression of ER, PR, HER2, CK5, CK14, EGFR, CD44, CD24, and ALDH1. The Chi square test or Fisher’s exact test was used to analyze the marker associations with clinical–pathologic tumor variables, molecular subtype and genetic subtype. Analyses of the association of overall survival (OS) with marker status were conducted using Kaplan–Meier plots and log-rank tests. The CD44+/CD24−/low and ALDH1+ phenotypes were identified in 16% and 15% of the familial breast cancer cases, respectively, and associated with high-tumor grade, a high-mitotic count, and component features of the medullary type of breast cancer. CD44+/CD24−/low and ALDH1 expression in this series were further associated with the basal-like molecular subtype and the CD44+/CD24−/low phenotype was independently associated with BRCA1 mutational status. The currently accepted breast CSCs markers are present in a minority of familial breast cancers. Whereas the presence of these markers is correlated with several poor prognostic features and the basal-like subtype of breast cancer, they do not predict OS.
Abkürzungen
ALDH1
Aldehyde dehydrogenase 1
BRCA1
Breast cancer 1, early onset
BRCA2
Breast cancer 2, early onset
CSC
Cancer stem cell
CD
Cluster designation
CK
Cytokeratin
DNA
Deoxyribonucleic acid
EGFR
Epidermal growth factor receptor
ER
Estrogen receptor
FFPE
Formalin fixed paraffin embedded
H&E
Hematoxylin and eosin
IHC
Immunohistochemistry
Negative
OS
Overall survival
+
Positive
PR
Progesterone receptor
RNA
Ribonucleic acid
TMA
Tissue microarray
WHO
World Health Organisation

Introduction

Breast cancer is a highly heterogenous disease at the morphologic and molecular level, with at least four main molecular subtypes described, including luminal (divided into luminal A and B), HER2 over-expressing, and basal-like [13]. More recently additional molecular subtypes have been indentified including claudin-low and molecular apocrine [46]. Each of these subtypes has characteristic morphologic, immunophenotypic, and prognostic features. BRCA1-associated breast cancers have been shown to be enriched with tumors of the basal-like subtype, whereas BRCA2-associated tumors and familial non-BRCA1-2 tumors are more likely to be of the luminal subtypes [711].
The normal epithelium of the breast has been demonstrated to be organized in a cellular hierarchy with an ER-negative (−) stem cell giving rise to ER-positive (+) and ER-negative progenitors, which ultimately give rise to fully differentiated functional luminal and myoepithelial/basal epithelium [12, 13]. It has been suggested that the different molecular subtypes of breast cancer arise from the transformation of different stem or progenitor cell populations which retain, or acquire as a consequence of the transformation process some or all of functional characteristics of normal stem cells [14]. These characteristics include limitless self-renewal capabilities, which drives tumorigenesis, and the ability to differentiate (albeit aberrantly) leading to morphologic tumor heterogeneity. There is also evidence to suggest that it is the CSC population that mediates metastases and can evade the effects of chemotherapy and radiation therapy thus promoting recurrence and relapse [1522].
A number of markers have been proposed that enrich for the identification of breast CSCs including CD44 in combination with low or absent expression of CD24 (known as the CD44+/CD24−/low phenotype) [23, 24] and Aldehyde dehydrogenase 1 (ALDH1) [25]. CD44 and CD24 are both adhesion molecules that play major roles in cell–cell and cell–extracellular matrix (ECM) interactions. CD44 is a Class I transmembrane glycoprotein that serves as the primary receptor for hyaluronan [26] and binds other ECM components, such as collagen, laminin, and fibronectin. CD44 exists in different splicing variants; some of these variants have been reported to promote growth, survival, invasion, and metastatic properties in breast cancer cells [2730]. However, other studies on the role of CD44 in breast cancer have shown opposite effects, suggesting that CD44’s function in breast cancer is context-dependent (reviewed [31]). CD24 is a small cell-surface glycoprotein that binds P-selectin, an adhesion receptor on platelets and endothelial cells [32]. In addition, CD24 promotes binding to fibronectin, collagen, and laminin and in agreement with these functions has been shown to promote adhesion, migration, and metastasis, and to associate with markers of poor prognosis in breast cancer [3234]. ALDH1 is a detoxifying enzyme responsible for the oxidation of intracellular aldehydes [35] that plays a role in early differentiation of stem cells by promoting the formation of retinoic acid [36]. In addition to preferential expression of ALDH1 in breast cancer cells with tumor initiating properties [25], retinoid signaling has been directly implicated in modulating breast cancer stem cell (CSC) differentiation [37].
In the present study, we examine the expression of the proposed breast CSC markers in a well-characterized collection of familial breast cancer cases. In addition, we investigated whether the expression of these markers is associated with any known clinical–pathologic tumor variables, molecular subtype or with patient survival.

Materials and methods

Study population

The study population included 58 BRCA1-associated, 64 BRCA2-associated, and 242 familial non-BRCA1/BRCA2 tumors from the Ontario Familial Breast Cancer Registry. All familial non-BRCA1/BRCA2 breast cancers were obtained from probands within the Breast Cancer Family Registry who met any of the following criteria for being at possible genetic risk of breast cancer; at least 1 first-degree relative with breast or ovarian cancer, at least 2 second-degree relatives with breast or ovarian cancer, diagnosis before age 26, male, multiple primaries or breast and ovarian cancer, at least 1 second-degree or third-degree relative with male breast cancer, multiple breast, or breast and ovarian primaries, or breast cancer before 26 years of age, or ovarian cancer before 60 years of age, Ashkenazi Jewish, or 3 first degree relatives in the family with breast, ovarian, colon, prostate or pancreatic cancer or sarcoma (with one diagnosed before age 50) but who tested negative for germline BRCA1 or BRCA2 mutations. Sporadic breast cancer cases from the registry were not available on TMAs.

Mutational analysis of BRCA1 and BRCA2

Testing for germline mutations in BRCA1 and BRCA2 was performed using an RNA/DNA-based protein truncation test with complementary 5′ sequencing, as previously described [38, 39], or by complete gene sequencing by Myriad Genetics. All mutations were confirmed by DNA sequencing. Mutations were classified as deleterious if they were protein-truncating, missense mutations (rare), or splice-site mutations as defined by the Breast Informatics Consortium (http://​research.​nhgri.​nih.​gov/​bic/​).

Pathology review

All tumors from the familial breast cancer cohort had a centralized pathology review performed by an expert breast pathologist using a standardized checklist form. The reviewing pathologist was unaware of the mutational status of the tumor at the time of review. Tumors were classified according to the WHO histologic classification of breast tumors and graded using the Nottingham histologic grading system [40, 41].

TMA construction

A suitable paraffin-embedded block of invasive tumor was chosen at the time of pathology review and the area of invasive tumor encircled for TMA construction. Two 0.6 mm cores of tissue were taken from the paraffin tumor block and used for TMA construction (Beecher Instruments, Sun Praire, WI) as previously described [7, 8]. Four μm sections were cut and immunohistochemical staining for ER, PR, HER2, CK5, CK14, EGFR, ALDH1, CD44, and CD24 was performed using methods as listed in Table 1. Microwave antigen retrieval was carried out in a Micromed T/T Mega Microwave Processing Lab Station (ESBE Scientific, Markham, Ontario, Canada). Sections were developed with diaminobenzidine tetrahydrochloride (DAB) and counterstained in Mayer’s hematoxylin.
Table 1
Summary of antibodies and their conditions of use
Antibody against
Clone
Source
Dilution
Pretreatment
ER
6F11
Vector
1/100
Microwave at 115 °C for 12 min in modified Tris–HCl buffer (pH 9)
PR
PgR1294
Dako
1/2000
Microwave at 115 °C for 12 min in modified Tris–HCl buffer (pH 9)
HER2
A0485
Dako
1/600
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)
CK5
XM26
Vector
1/400
Microwave at 115 °C for 12 min in modified Tris–HCl buffer (pH 9)
CK14
LL002
Vector
1/40
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)
EGFR
31G7
Invitrogen
1/35
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)
Ki67
MIB1
Dako
1/300
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)
ALDH1
44ALDH (ALDH1A1)
BD Biosciences
1/200
Microwave at 115 °C for 12 min in modified Tris–HCl buffer (pH 9)
CD44
DF1485
Dako
1/250
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)
CD24
SN3b
Neomarkers
1/45
Microwave at 115 °C for 12 min in modified citrate buffer (pH 6)

Interpretation and scoring of immunohistochemistry

Each of the immunohistochemical TMA-stained sections was scored using Allred’s scoring method [42], which adds scores for the intensity of staining (absent: 0, weak: 1, moderate: 2, and strong: 3 to the percentage of cells stained (none: 0, <1%: 1, 1–10%: 2, 11–33%: 3, 34–66%: 4, and 67–100%: 5 to yield a “raw” score of 0 or 2–8. Previously validated cut-offs for ER and PR were used (0, 2 = negative, 3–8 = positive) [43, 44]. Strong complete membranous staining was assessed for HER2 and the cut-off of >5 was used to indicate positivity [45]. For CK5, CK14, EGFR, CD44, and CD24 a score of ≥4 was considered positive, for ALDH1 a score of ≥5 was considered positive. The raw score data were reformatted using a TMA deconvoluter software program into a format suitable for statistical analysis [46]. The highest score from each TMA tumor pair was entered into the statistical analysis. Only the epithelial component of each TMA spot was scored for the markers indicated. Immunohistochemical results were recorded as unavailable when the tissue sections were washed off the slide, TMA cores contained no invasive tumor cells or when sections were uninterpretable due to tissue artifact.
Tumors were classified as luminal if they expressed ER or PR and were negative for HER2. Any tumor with a score of >5 for HER2, irrespective of the ER status was considered a HER2 over-expressing tumor and basal-like tumors were defined as ER, PR, and HER2 negative (triple negative) and positive for CK5 and/or CK14 and/or EGFR as previously described [7, 8, 47].

Statistical analysis

The Chi square test or Fisher’s exact test was used to analyze the marker associations with clinical–pathologic tumor variables, molecular subtype and genetic subtype. Analyses of the association of OS (overall survival) with marker status were conducted using Kaplan–Meier plots and log-rank tests. The follow up data were to the end of November 24, 2011. Excluding the patients lost to follow-up and those with deaths, the minimum follow-up time was 12 months after surgery and the median follow-up time was 148 months. Patient status on November 24, 2011, determined OS time and censoring status. All tests were two-sided. A test with a P-value < 0.05 was considered statistically significant. P-values were not adjusted for multiple testing. Statistical analysis of associations was performed using SAS 9.1 software (SAS Institute, Inc.). Survival curves were plotted using R statistical software, version 2.15.0 (http://​www.​r-project.​org/​).

Results

CD44+/CD24−/low

Two hundred and sixty two (262) cases had results for both CD44 and CD24, of which 41 (16%) had a CD44+/CD24−/low phenotype (Table 2; Fig. 1a–d). When compared with all other combinations of CD44 and CD24 expression, the CD44+/CD24−/low phenotype was positively associated with high-tumor grade (p = 0.03), a high-mitotic score (p = 0.003), margin circumscription (p = 0.0009), a moderate tumor lymphocytic infiltrate (p = 0.01), and absent lympho-vascular space invasion (p = 0.008). In addition there was a statistically non-significant trend in association between the CD44+/CD24−/low phenotype and the lack of lymph-node metastases (p = 0.09), syncytial tumor growth pattern (p = 0.06) and young age at diagnosis (p = 0.06). No association was detected between the CD44+/CD24−/low phenotype and tumor size, or tumor type.
Table 2
Association between the CD44+/CD24−/low phenotype and tumor morphologic characteristics
Tumor characteristic
CD44+/CD24−/low (n = 41)
Other combinations of CD44 and CD24 (n = 221)
p value
n
%
n
%
Grade
 III
31
75.6
118
53.6
0.03
 II
6
14.6
73
33.2
 I
4
9.8
29
13.2
Size (mm)
 0–20
24
58.5
124
56.6
0.97
 20–50
15
36.6
83
37.9
 >50
2
4.9
12
5.5
Type
 Invasive ductal
40
97.6
204
92.7
0.22
 Invasive lobular
1
2.4
13
5.9
 Other
0
0.0
3
1.4
Lympho-vascular space invasion
 Positive
9
22.5
99
45.0
0.008
 Negative
31
77.5
121
55.0
Mitotic score
 1
9
22.0
82
37.3
0.003
 2
3
7.3
46
20.9
 3
29
70.7
92
41.8
Number of LN positive
 0
26
68.4
109
52.9
0.09
 1–3
11
29.0
59
28.6
 4–9
1
2.6
30
14.6
 ≥10
0
0.0
8
3.9
Margin circumscription
 Positive
19
47.5
49
22.4
0.0009
 Negative
21
52.5
170
77.6
Syncytial growth
 Positive
7
17.5
16
73
0.06
 Negative
33
82.5
203
92.7
Age at diagnosis (years)
 <40
21
51.2
72
32.6
0.06
 40–50
12
29.3
103
46.6
 >50
8
19.5
46
20.8
Lymphocytic infiltrate
 Absent (0)
3
7.5
68
31.1
0.01
 Weak (1)
6
15.0
17
7.8
 Mild (4)
19
47.5
91
41.5
 Moderate (5)
12
30.0
43
19.6
LN lymph node
In the 41 CD44+/CD24−/low cases a molecular phenotype was assignable for 33 tumors (Table 3), 16 (48.5%) of which were basal, 1 (3%) was HER2 overexpressing and 16 (48.5%) were luminal. In comparison to all other combinations of CD44 and CD24 expression, tumors with a CD44+/CD24−/low phenotype were more likely to belong to the basal-like molecular subtype (48.5 vs. 22.2%; p = 0.0034).
Table 3
Association between the CD44+/CD24−/low phenotype and tumor molecular subtype
CD44/CD24 status
Molecular subtype
p value
Basal
HER-2
Luminal
n
%
n
%
n
%
CD44+/CD24−/low
16
48.5
1
3.0
16
48.5
0.0034
Other combinations of CD44 and CD24
44
22.2
28
14.1
126
63.7
Of the 41 CD44+/CD24−/low cases, 11 (27%) were BRCA1-associated tumors, 7 (17%) were BRCA2-associated tumors, and the remainder 23 (56%) were from non-BRCA1/BRCA2 tumors (Table 4). When compared to all other combinations of CD44 and CD24 expression, a CD44+/CD24−/low phenotype was more likely to be associated with tumors arising in BRCA1 germline mutation carriers than non-BRCA1 mutation carriers (26.8 vs. 12.7%; p = 0.02). However, when the analysis was restricted to basal tumors only (Table 5), there was no statistical difference in the incidence of CD44+/CD24−/low expression in BRCA1-associated basal tumors and non-BRCA1-associated basal tumors.
Table 4
Association between the CD44+/CD24−/low phenotype and tumor genetic subgroup
CD44/CD24 status
Tumor subgroup
p value
BRCA1
BRCA2
Familial non-BRCA1/2
n
%
n
%
n
%
CD44+/CD24−/low
11
26.8
7
17.1
23
56.1
0.0644
Other
28
12.7
47
21.3
146
66.0
Table 5
Association between the CD44+/CD24−/low phenotype and tumor genetic subgroup within the basal-like molecular subtype
CD44/CD24 status
Tumor subgroup
p value
BRCA1-basal
Non-BRCA1-basal
n
%
n
%
CD44+/CD24−/low
6
37.5
10
62.5
0.69
Other combinations of CD44 and CD24
19
43.2
25
56.8

ALDH1

ALDH1 was expressed in 39 of 255 (15%) tumors (Table 6; Fig. 2a, b). The expression of ALDH1 was positively associated with high-tumor grade (p = 0.003), large tumor size (p = 0.009), high-mitotic score (p = 0.05), a syncytial growth pattern (p < 0.0001), a moderate tumor lymphocytic infiltrate (p = 0.002) and younger age at diagnosis (p = 0.02). No statistically significant association was detected between ALDH1 expression and tumor type, lympho-vascular space invasion, or lymph-node status.
Table 6
Association between ALDH1 expression and tumor morphologic characteristics
Tumor characteristic
ALDH1+ (n = 39)
ALDH1− (n = 216)
p value
n
%
n
%
Grade
 III
30
79.0
112
52.1
0.0033
 II
8
21.0
69
32.1
 I
0
0.0
34
15.8
Size (mm)
 0–20
14
36.9
133
62.2
0.0099
 20–50
20
52.6
72
33.6
 >50
4
10.5
9
4.2
Type
 Invasive ductal
36
94.8
198
92.1
1.0000
 Invasive lobular
1
2.6
11
5.1
 Other
1
2.6
6
2.8
Lympho-vascular space invasion
 Positive
16
42.1
87
40.7
0.8668
 Negative
22
57.9
127
59.3
Mitotic score
 1
8
21.0
85
39.5
0.0549
 2
6
15.8
37
17.2
 3
24
63.2
93
43.3
Number of LN positive
 0
20
57.1
118
59.0
0.7104
 1–3
8
22.9
53
26.5
 4–9
6
17.1
21
10.5
 ≥10
1
2.9
8
4.0
Margin circumscription
 Positive
15
39.5
53
24.9
0.0623
 Negative
23
60.5
160
75.1
Syncytial growth
 Positive
10
26.3
13
6.1
<0.0001
 Negative
28
73.7
200
93.9
Age at diagnosis (years)
 <40
21
53.9
69
32.0
0.0214
 40–50
10
25.6
99
45.8
 >50
8
20.5
48
22.2
Lymphocytic infiltrate
 Absent
4
10.5
64
30.0
0.0025
 Weak
7
18.4
16
7.5
 Mild
13
34.2
93
43.7
 Moderate
14
36.9
40
18.8
LN lymph node
A molecular subtype was assignable in 33 of 39 ALDH1 positive tumors, 16 (48.5%) of which were basal, 3 (9%) were HER2 overexpressing and 14 (42.5%) were luminal (Table 7). When compared to tumors lacking ALDH1 expression, ALDH1 positive tumors were more commonly basal-like (48.5 vs. 22.3%; p = 0.007).
Table 7
Association between ALDH1 expression and tumor molecular subtype
ALDH1 status
Molecular subtype
p value
Basal
HER-2
Luminal
n
%
n
%
n
%
Positive
16
48.5
3
9.1
14
42.4
0.007
Negative
43
22.3
24
12.4
126
65.3
Of the 39 ALDH1 expressing tumors, 9 (23%) were from BRCA1 germline mutation carriers, 9 (23%) were from BRCA2 germline mutation carriers, and 21 (54%) were from non-BRCA1/BRCA2 patients (Table 8). There was no statistically significant association between ALDH1 expression and BRCA1 mutational status (23.1 vs. 14.3%; p = 0.17), even when the analysis was restricted to BRCA1 basal-like tumors only (data not shown).
Table 8
Association between ALDH1 expression and tumor genetic subgroup
ALDH1 status
Tumor subgroup
p value
BRCA1
BRCA2
Control
n
%
n
%
n
%
Positive
9
23.1
9
23.1
21
53.8
0.2869
Negative
31
14.4
43
19.9
142
65.7

CD44+/CD24−/low/ALDH1+

For the familial breast cancer series the combined CD44+/CD24−/low/ALDH1+ phenotype was expressed in 6 of 230 tumors (data not shown) and associated with a high-mitotic score (p = 0.04), high-mitotic count (p = 0.03), and a syncytial growth pattern (p = 0.01). There was a non-statistically significant trend toward an association with tumor size (p = 0.09), lympho-vascular space invasion (p = 0.08), young age at diagnosis (p = 0.08), and tumor lymphocytic infiltrate (p = 0.08). No association was found between the expression of these combined markers and tumor grade (p = 0.21), tumor type (p = 1.0), lymph-node involvement (p = 0.42) or margin circumscription (p = 0.36).
Only 6 cases expressed a combined CD44+/CD24−/low/ALDH1+ phenotype and while this number of tumors is too few to perform a robust analysis we did observe that 2 (33%) were basal-like tumors and the remaining 4 (67%) were luminal tumors. In these 6 tumors, 3 (50%) were from BRCA1 germline mutation carriers, none (0%) were from BRCA2 germline mutation carriers and the remaining 3 (50%) were from non-BRCA1/BRCA2 mutation carriers. When compared to all other combinations of CD44, CD24, and ALDH1 expression, tumors with a CD44+/CD24−/low/ALDH1+ phenotype were more likely to be associated with BRCA1 germline mutation carriers than non-mutation carriers (data not shown). On analysis of the tumors with a basal-like molecular subtype only (data not shown), there was no significant difference in CD44+/CD24−/low/ALDH1+ expression between those tumors with and without a BRCA1 germline mutation.

Survival

There was a non-significant trend toward better survival for the group with CD44+/CD24−/low compared to the group with other combinations of CD44 and CD24 (Fig. 3). There was no difference in survival between patients with tumors positive for ALDH1 and tumors negative for this marker (Fig. 4).

Discussion

There is an increasing evidence that many tumors including breast cancers may be driven by a subpopulation of cells that display stem cell properties, so called CSCs or tumor initiating cells. Markers have been identified that when used alone or in combination enrich for functional CSCs, as defined by their ability to selectively initiate tumors in immunocompromised mice upon serial passage, a demonstration of self renewal, together with the ability to form tumors that are heterogeneous at the cellular level similar to the originating tumor, illustrative of the CSC’s ability to differentiate [48]. These markers include CD44+/CD24−/low and ALDH1, originally identified by the sorting of cells from fresh tumors or effusions using flow cytometry or an enzymatic assay [23, 25]. Unfortunately fresh tumor samples are not routinely available for all breast cancer patients and tumor effusions manifest at a relatively late stage of the disease process and may not be representative of the primary tumor. In order to investigate whether CSCs could represent either prognostic or predictive biomarkers an alterative approach to their identification must be sought, preferably in formalin fixed paraffin embedded (FFPE) tumor material which represents the bulk of patient tumor samples and clinical trial archives. In this study we have used immunohistochemical (IHC) expression of CD44+/CD24−/low and ALDH1 as surrogate markers for breast CSCs and sought to correlate their expression alone and in combination with clinical–pathologic tumor features, breast cancer molecular subtypes, germline gene mutations, and ultimately patient outcome.
Our observations suggest that only a minority of the tumors examined contained cells expressing the breast CSC phenotypes CD44+/CD24−/low(15%), ALDH1 (16%) or both combined CD44+/CD24−/low/ALDH1+ (<1%). Other investigators employing IHC methods to identify these phenotypes have reported a wide variance in the percentage of primary breast tumors that express these phenotypes: 20–60% of tumors exhibit some cells with a CD44+/CD24−/low phenotype [4955], whereas 7–70% of tumors examined expressed ALDH1 [22, 25, 52, 54, 5661]. These differences may reflect differences in the antibodies employed, or the tumor populations examined e.g., familial vs sporadic, or the scoring cut points applied (for example, some studies considered tumors with as few as one cell positive for the markers to be positive [50, 59] whereas others like this study have required a minimum of 10% of tumor cells to express the marker in question for the tumor to be considered positive [52, 54]). Alternatively, perhaps CSCs are phenotypically more diverse and the two phenotypes we have examined may not capture all possible breast CSCs. Wright et al. [62] in an examination of CSCs from transgenic mice engineered to be deficient in BRCA1 demonstrated that some tumors contained CSCs with a CD44+/CD24−/low phenotype, whereas other tumors contained CSC characterized by CD133 expression. Other markers identified as putative breast CSC markers include CK5, EGFR, EpCAM, and CD49f [63, 64].
Traditionally in breast cancer a number of clinical–pathologic tumor characteristics are associated with poor prognosis and include; younger age at diagnosis, large tumor size, lymph-node involvement, high-tumor grade, lympho-vascular space invasion (LVSI), negative hormonal receptor status, and HER2 over-expression [65, 66]. Patients with tumors displaying some or all of these features are considered at increased risk for relapse and death from breast cancer when compared to patients with tumors lacking these features. In this study, we demonstrated a positive association between the presence of cells with a CSC phenotype (CD44+/CD24−/low or ALDH1 positive or both) and many of these adverse prognostic features including high-tumor grade, large tumor size, and younger age at diagnosis. A number of other studies have reported similar associations between the presence of CSCs and adverse prognostic features [49, 52, 53]. Interestingly, despite the association with some adverse prognostic factors we were unable to demonstrate an association between CD44+/CD24−/low or ALDH1 expression and breast cancer outcome. We are not alone in this observation, in 2 of 4 other studies where the expression of CD44+/CD24−/low was analyzed in relation to outcome no association was observed [51, 54], in the third study an association between CD44+/CD24−/low and outcome was significant on univariate analyses only [53], whereas in the fourth an inverse relationship between CD44+/CD24−/low expression and survival was reported [55]. The expression of ALDH1 has been correlated with poor patient prognosis in some but not all studies [25, 54, 57, 58, 61, 67]. In two studies the expression of ALDH1 was found to be an independent prognostic variable after multivariate analyses [25, 57]. However, similar to our study Ricardo et al. [54] and Resetkova et al. [61] failed to demonstrate an association between ALDH1 expression and outcome. The lack of an association between these markers and patient survival in a number of studies may suggest that the presence of cells with a stem cell phenotype is not a prognostic marker or alternatively that the identification of these markers by immunohistochemistry may not accurately identify the functional CSC population within a tumor.
In our study, the CD44+/CD24−/low and ALDH1 phenotypes were positively associated with the component features of medullary-type breast cancer; namely prominent lymphocytic infiltrate, pushing tumor margins and syncytial growth pattern [68]. Medullary cancer is a special subtype of breast cancer that occurs in 1–5% of all cases, these cancers are ER, PR, and HER2 negative and characteristically high grade [40, 69]. Furthermore, they have been demonstrated to cluster with either basal-like or claudin-low molecular subtypes of breast cancer and to be more commonly represented in tumors of BRCA1 mutation carriers [3, 5, 70, 71]. Despite these seemingly adverse morphologic and molecular associations medullary-type cancers are associated with a better prognosis than non-medullary grade III tumors a fact that may result from the prominent host lymphocytic response that characterizes these tumors [69]. A prominent tumor lymphocytic infiltrate has been demonstrated to be a good prognostic factor in ER-negative breast cancer and basal-like breast cancers specifically [7274]. It is plausible that the presence of an “anti-tumor” immune response in the tumor stroma may mitigate the effects of the increase in CSCs present in these tumor types.
The basal-like subtype of breast cancer is a molecular subtype that was originally discovered through gene expression profiling studies [13, 75]. This subtype if predominantly triple negative (ER, PR, and HER2 negative) and associated, at least in the short-term, with a worse prognosis than ER-positive luminal-type tumors [7678]. Breast tumors from patients with BRCA1 germline mutations are enriched for this subtype [3, 7, 9]. In this study, we demonstrate that both CSCs expressing either the CD44+/CD24−/low or ALDH1 positive phenotype are more commonly found in basal-like tumors than any other molecular subtype examined. Furthermore, we demonstrated  a positive association between BRCA1 mutational status and the CD44+/CD24−/low phenotype. BRCA1 is believed to be a regulator of breast stem cell fate and is required for mammary epithelial cell differentiation [79, 80]. Specifically, BRCA1 is required for the differentiation of ER− luminal progenitor cells and in its absence, such as in the epithelium of BRCA1 mutation carriers, the transformed luminal progenitors are “driven” toward a basal cell fate (expressing CK5), hence the predominance of the basal-like tumor phenotype among BRCA1 mutation carriers [64, 81, 82]. Sporadic basal-like breast cancer arising in patients without germline BRCA1 mutations are often deficient in functional BRCA1 protein resulting in a similar pathway for the development of sporadic and BRCA1-associated basal-like breast cancer and hence a similar phenotype and CSC expression [83]. Honeth et al. [50] profiled 17 BRCA1-associated breast cancers for the CD44+/CD24−/low phenotype and found that 94% of their BRCA1-associated tumors expressed this phenotype as did 63% of the sporadic basal-like breast cancer included in the study. Heerma van Voss et al. [56] demonstrated that ALDH1 was an independent predictor of BRCA1 mutational status, whereas in our larger cohort of BRCA1-associated tumors ALDH1 was not associated with BRCA1 status but rather with the basal-like subtype only.
In our study, a very small fraction of tumors examined (6 of 230 tumors or 0.03%) expressed both CSC phenotypes. Unfortunately, this small number of cases precludes robust statistical analysis but another study by Rimm et al using AQUA technology on the Yale breast cancer cohort showed that 5.5% of breast tumors examined contained cells that co-expressed both CD44 and ALDH1 and these tumors were associated with a high breast cancer-specific mortality [67]. This is in agreement with observations by Ginestier et al. [25], who have shown that tumor cells expressing both phenotypes are highly tumorigenic with the capacity to generate tumors from as few as 20 cells in vivo.
In conclusion, we have demonstrated that CSCs as defined by the expression of CD44+/CD24−/low and/or ALDH1 are present in a minority of familial breast cancer cases. The expression of these CSC phenotypes is associated with a number of adverse prognostic clinical–pathologic features but not with overall survival. In addition, we have demonstrated that the expression of CD44+/CD24−/low and/or ALDH1 is more common in basal-like tumors and that there is an association between BRCA1 mutational status and CD44+/CD24−/low expression. 

Acknowledgments

This work was supported by the Ontario Institute for Cancer Research, Award 1A-008, the Canadian Breast Cancer Foundation and the US National Cancer Institute, National Institutes of Health under RFA # CA-06-503 and through cooperative agreements with members of the Breast Cancer Family Registry and Principal Investigators, including Cancer Care Ontario (U01 CA69467). The content of this manuscript does not necessarily reflect the views or policies of the National Cancer Institute or any of the collaborating centers in the BCFR, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government or the BCFR.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Perou CM, Sorlie T, Eisen MB et al (2000) Molecular portraits of human breast tumours. Nature 406:747–752PubMedCrossRef Perou CM, Sorlie T, Eisen MB et al (2000) Molecular portraits of human breast tumours. Nature 406:747–752PubMedCrossRef
2.
Zurück zum Zitat Sorlie T, Perou CM, Tibshirani R et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98:10869–10874PubMedCrossRef Sorlie T, Perou CM, Tibshirani R et al (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98:10869–10874PubMedCrossRef
3.
Zurück zum Zitat Sorlie T, Tibshirani R, Parker J et al (2003) Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100:8418–8423CrossRef Sorlie T, Tibshirani R, Parker J et al (2003) Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci U S A 100:8418–8423CrossRef
4.
Zurück zum Zitat Herschkowitz JI, Simin K, Weigman VJ et al (2007) Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol 8:R76PubMedCrossRef Herschkowitz JI, Simin K, Weigman VJ et al (2007) Identification of conserved gene expression features between murine mammary carcinoma models and human breast tumors. Genome Biol 8:R76PubMedCrossRef
5.
Zurück zum Zitat Prat A, Parker JS, Karginova O et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12:R68PubMedCrossRef Prat A, Parker JS, Karginova O et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12:R68PubMedCrossRef
6.
Zurück zum Zitat Farmer P, Bonnefoi H, Becette V et al (2005) Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 24:4660–4671PubMedCrossRef Farmer P, Bonnefoi H, Becette V et al (2005) Identification of molecular apocrine breast tumours by microarray analysis. Oncogene 24:4660–4671PubMedCrossRef
7.
Zurück zum Zitat Mulligan AM, Pinnaduwage D, Bane AL et al (2010) CK8/18 expression, the basal phenotype, and family history in identifying BRCA1-associated breast cancer in the Ontario site of the Breast Cancer Family Registry. Cancer 117(7):1350–1359PubMedCrossRef Mulligan AM, Pinnaduwage D, Bane AL et al (2010) CK8/18 expression, the basal phenotype, and family history in identifying BRCA1-associated breast cancer in the Ontario site of the Breast Cancer Family Registry. Cancer 117(7):1350–1359PubMedCrossRef
8.
Zurück zum Zitat Bane AL, Beck JC, Bleiweiss I et al (2007) BRCA2 mutation-associated breast cancers exhibit a distinguishing phenotype based on morphology and molecular profiles from tissue microarrays. Am J Surg Pathol 31:121–128PubMedCrossRef Bane AL, Beck JC, Bleiweiss I et al (2007) BRCA2 mutation-associated breast cancers exhibit a distinguishing phenotype based on morphology and molecular profiles from tissue microarrays. Am J Surg Pathol 31:121–128PubMedCrossRef
9.
Zurück zum Zitat Lakhani SR, Reis-Filho JS, Fulford L et al (2005) Prediction of BRCA1 status in patients with breast cancer using estrogen receptor and basal phenotype. Clin Cancer Res 11:5175–5180PubMedCrossRef Lakhani SR, Reis-Filho JS, Fulford L et al (2005) Prediction of BRCA1 status in patients with breast cancer using estrogen receptor and basal phenotype. Clin Cancer Res 11:5175–5180PubMedCrossRef
10.
Zurück zum Zitat Palacios J, Honrado E, Osorio A et al (2005) Phenotypic characterization of BRCA1 and BRCA2 tumors based in a tissue microarray study with 37 immunohistochemical markers. Breast Cancer Res Treat 90:5–14PubMedCrossRef Palacios J, Honrado E, Osorio A et al (2005) Phenotypic characterization of BRCA1 and BRCA2 tumors based in a tissue microarray study with 37 immunohistochemical markers. Breast Cancer Res Treat 90:5–14PubMedCrossRef
11.
Zurück zum Zitat Palacios J, Honrado E, Osorio A et al (2003) Immunohistochemical characteristics defined by tissue microarray of hereditary breast cancer not attributable to BRCA1 or BRCA2 mutations: differences from breast carcinomas arising in BRCA1 and BRCA2 mutation carriers. Clin Cancer Res 9:3606–3614PubMed Palacios J, Honrado E, Osorio A et al (2003) Immunohistochemical characteristics defined by tissue microarray of hereditary breast cancer not attributable to BRCA1 or BRCA2 mutations: differences from breast carcinomas arising in BRCA1 and BRCA2 mutation carriers. Clin Cancer Res 9:3606–3614PubMed
12.
Zurück zum Zitat Booth BW, Smith GH (2006) Estrogen receptor-alpha and progesterone receptor are expressed in label-retaining mammary epithelial cells that divide asymmetrically and retain their template DNA strands. Breast Cancer Res 8:R49PubMedCrossRef Booth BW, Smith GH (2006) Estrogen receptor-alpha and progesterone receptor are expressed in label-retaining mammary epithelial cells that divide asymmetrically and retain their template DNA strands. Breast Cancer Res 8:R49PubMedCrossRef
13.
Zurück zum Zitat Sleeman KE, Kendrick H, Robertson D et al (2007) Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol 176:19–26PubMedCrossRef Sleeman KE, Kendrick H, Robertson D et al (2007) Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol 176:19–26PubMedCrossRef
14.
Zurück zum Zitat Dontu G, Al-Hajj M, Abdallah WM et al (2003) Stem cells in normal breast development and breast cancer. Cell Prolif 36(Suppl 1):59–72PubMedCrossRef Dontu G, Al-Hajj M, Abdallah WM et al (2003) Stem cells in normal breast development and breast cancer. Cell Prolif 36(Suppl 1):59–72PubMedCrossRef
15.
Zurück zum Zitat Theodoropoulos PA, Polioudaki H, Agelaki S et al (2009) Circulating tumor cells with a putative stem cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett 288(1):99–106 Theodoropoulos PA, Polioudaki H, Agelaki S et al (2009) Circulating tumor cells with a putative stem cell phenotype in peripheral blood of patients with breast cancer. Cancer Lett 288(1):99–106
16.
Zurück zum Zitat Aktas B, Tewes M, Fehm T et al (2009) Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res 11:R46PubMedCrossRef Aktas B, Tewes M, Fehm T et al (2009) Stem cell and epithelial-mesenchymal transition markers are frequently overexpressed in circulating tumor cells of metastatic breast cancer patients. Breast Cancer Res 11:R46PubMedCrossRef
17.
Zurück zum Zitat Lu J, Fan T, Zhao Q et al (2009) Isolation of circulating epithelial and tumor progenitor cells with an invasive phenotype from breast cancer patients. Int J Cancer 126(3):669–683 Lu J, Fan T, Zhao Q et al (2009) Isolation of circulating epithelial and tumor progenitor cells with an invasive phenotype from breast cancer patients. Int J Cancer 126(3):669–683
18.
Zurück zum Zitat Balic M, Lin H, Young L et al (2006) Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res 12:5615–5621PubMedCrossRef Balic M, Lin H, Young L et al (2006) Most early disseminated cancer cells detected in bone marrow of breast cancer patients have a putative breast cancer stem cell phenotype. Clin Cancer Res 12:5615–5621PubMedCrossRef
19.
Zurück zum Zitat Phillips TM, McBride WH, Pajonk F (2006) The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 98:1777–1785PubMedCrossRef Phillips TM, McBride WH, Pajonk F (2006) The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst 98:1777–1785PubMedCrossRef
20.
Zurück zum Zitat Diehn M, Cho RW, Lobo NA et al (2009) Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458:780–783PubMedCrossRef Diehn M, Cho RW, Lobo NA et al (2009) Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458:780–783PubMedCrossRef
21.
Zurück zum Zitat Li X, Lewis MT, Huang J et al (2008) Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100:672–679PubMedCrossRef Li X, Lewis MT, Huang J et al (2008) Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J Natl Cancer Inst 100:672–679PubMedCrossRef
22.
Zurück zum Zitat Tanei T, Morimoto K, Shimazu K et al (2009) Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res 15:4234–4241PubMedCrossRef Tanei T, Morimoto K, Shimazu K et al (2009) Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clin Cancer Res 15:4234–4241PubMedCrossRef
23.
Zurück zum Zitat Al-Hajj M, Wicha MS, Benito-Hernandez A et al (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100:3983–3988PubMedCrossRef Al-Hajj M, Wicha MS, Benito-Hernandez A et al (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 100:3983–3988PubMedCrossRef
24.
Zurück zum Zitat Ponti D, Costa A, Zaffaroni N et al (2005) Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 65:5506–5511PubMedCrossRef Ponti D, Costa A, Zaffaroni N et al (2005) Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res 65:5506–5511PubMedCrossRef
25.
Zurück zum Zitat Ginestier C, Hur MH, Charafe-Jauffret E et al (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1:555–567PubMedCrossRef Ginestier C, Hur MH, Charafe-Jauffret E et al (2007) ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1:555–567PubMedCrossRef
26.
Zurück zum Zitat Aruffo A, Stamenkovic I, Melnick M et al (1990) CD44 is the principal cell surface receptor for hyaluronate. Cell 61:1303–1313PubMedCrossRef Aruffo A, Stamenkovic I, Melnick M et al (1990) CD44 is the principal cell surface receptor for hyaluronate. Cell 61:1303–1313PubMedCrossRef
27.
Zurück zum Zitat Gunthert U, Hofmann M, Rudy W et al (1991) A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 65:13–24PubMedCrossRef Gunthert U, Hofmann M, Rudy W et al (1991) A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 65:13–24PubMedCrossRef
28.
Zurück zum Zitat Herrlich P, Morrison H, Sleeman J et al (2000) CD44 acts both as a growth- and invasiveness-promoting molecule and as a tumor-suppressing cofactor. Ann N Y Acad Sci 910:106–118 discussion 118-20PubMedCrossRef Herrlich P, Morrison H, Sleeman J et al (2000) CD44 acts both as a growth- and invasiveness-promoting molecule and as a tumor-suppressing cofactor. Ann N Y Acad Sci 910:106–118 discussion 118-20PubMedCrossRef
29.
Zurück zum Zitat Bourguignon LY, Wong G, Earle C et al (2010) Hyaluronan–CD44 interaction promotes c-Src-mediated twist signaling, microRNA-10b expression, and RhoA/RhoC up-regulation, leading to Rho-kinase-associated cytoskeleton activation and breast tumor cell invasion. J Biol Chem 285:36721–36735PubMedCrossRef Bourguignon LY, Wong G, Earle C et al (2010) Hyaluronan–CD44 interaction promotes c-Src-mediated twist signaling, microRNA-10b expression, and RhoA/RhoC up-regulation, leading to Rho-kinase-associated cytoskeleton activation and breast tumor cell invasion. J Biol Chem 285:36721–36735PubMedCrossRef
30.
Zurück zum Zitat Brown RL, Reinke LM, Damerow MS et al (2011) CD44 splice isoform switching in human and mouse epithelium is essential for epithelial–mesenchymal transition and breast cancer progression. J Clin Invest 121:1064–1074PubMedCrossRef Brown RL, Reinke LM, Damerow MS et al (2011) CD44 splice isoform switching in human and mouse epithelium is essential for epithelial–mesenchymal transition and breast cancer progression. J Clin Invest 121:1064–1074PubMedCrossRef
31.
Zurück zum Zitat Louderbough JM, Schroeder JA (2011) Understanding the dual nature of CD44 in breast cancer progression. Mol Cancer Res 9:1573–1586PubMedCrossRef Louderbough JM, Schroeder JA (2011) Understanding the dual nature of CD44 in breast cancer progression. Mol Cancer Res 9:1573–1586PubMedCrossRef
32.
Zurück zum Zitat Fogel M, Friederichs J, Zeller Y et al (1999) CD24 is a marker for human breast carcinoma. Cancer Lett 143:87–94PubMedCrossRef Fogel M, Friederichs J, Zeller Y et al (1999) CD24 is a marker for human breast carcinoma. Cancer Lett 143:87–94PubMedCrossRef
33.
Zurück zum Zitat Baumann P, Cremers N, Kroese F et al (2005) CD24 expression causes the acquisition of multiple cellular properties associated with tumor growth and metastasis. Cancer Res 65:10783–10793PubMedCrossRef Baumann P, Cremers N, Kroese F et al (2005) CD24 expression causes the acquisition of multiple cellular properties associated with tumor growth and metastasis. Cancer Res 65:10783–10793PubMedCrossRef
34.
Zurück zum Zitat Athanassiadou P, Grapsa D, Gonidi M et al (2009) CD24 expression has a prognostic impact in breast carcinoma. Pathol Res Pract 205:524–533PubMedCrossRef Athanassiadou P, Grapsa D, Gonidi M et al (2009) CD24 expression has a prognostic impact in breast carcinoma. Pathol Res Pract 205:524–533PubMedCrossRef
35.
Zurück zum Zitat Sophos NA, Vasiliou V (2003) Aldehyde dehydrogenase gene superfamily: the 2002 update. Chem Biol Interact 143–144:5–22PubMedCrossRef Sophos NA, Vasiliou V (2003) Aldehyde dehydrogenase gene superfamily: the 2002 update. Chem Biol Interact 143–144:5–22PubMedCrossRef
36.
Zurück zum Zitat Chute JP, Muramoto GG, Whitesides J et al (2006) Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci U S A 103:11707–11712PubMedCrossRef Chute JP, Muramoto GG, Whitesides J et al (2006) Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci U S A 103:11707–11712PubMedCrossRef
37.
Zurück zum Zitat Ginestier C, Wicinski J, Cervera N et al (2009) Retinoid signaling regulates breast cancer stem cell differentiation. Cell Cycle 8:3297–3302PubMedCrossRef Ginestier C, Wicinski J, Cervera N et al (2009) Retinoid signaling regulates breast cancer stem cell differentiation. Cell Cycle 8:3297–3302PubMedCrossRef
38.
Zurück zum Zitat Andrulis IL, Anton-Culver H, Beck J et al (2002) Comparison of DNA- and RNA-based methods for detection of truncating BRCA1 mutations. Hum Mutat 20:65–73PubMedCrossRef Andrulis IL, Anton-Culver H, Beck J et al (2002) Comparison of DNA- and RNA-based methods for detection of truncating BRCA1 mutations. Hum Mutat 20:65–73PubMedCrossRef
39.
Zurück zum Zitat Youil R, Kemper BW, Cotton RG (1995) Screening for mutations by enzyme mismatch cleavage with T4 endonuclease VII. Proc Natl Acad Sci U S A 92:87–91PubMedCrossRef Youil R, Kemper BW, Cotton RG (1995) Screening for mutations by enzyme mismatch cleavage with T4 endonuclease VII. Proc Natl Acad Sci U S A 92:87–91PubMedCrossRef
40.
Zurück zum Zitat Tavassoli T, Devilee P (2003) Pathology and genetics. Tumours of the breast and female genital organs. IARC Press, Lyon Tavassoli T, Devilee P (2003) Pathology and genetics. Tumours of the breast and female genital organs. IARC Press, Lyon
41.
Zurück zum Zitat Elston CW, Ellis IO (1991) Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410PubMedCrossRef Elston CW, Ellis IO (1991) Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19:403–410PubMedCrossRef
42.
Zurück zum Zitat Allred DC, Harvey JM, Berardo M et al (1998) Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168PubMed Allred DC, Harvey JM, Berardo M et al (1998) Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol 11:155–168PubMed
43.
Zurück zum Zitat Harvey JM, Clark GM, Osborne CK et al (1999) Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17:1474–1481PubMed Harvey JM, Clark GM, Osborne CK et al (1999) Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer. J Clin Oncol 17:1474–1481PubMed
44.
Zurück zum Zitat Mohsin SK, Weiss H, Havighurst T et al (2004) Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol 17:1545–1554PubMedCrossRef Mohsin SK, Weiss H, Havighurst T et al (2004) Progesterone receptor by immunohistochemistry and clinical outcome in breast cancer: a validation study. Mod Pathol 17:1545–1554PubMedCrossRef
45.
Zurück zum Zitat Wolff AC, Hammond ME, Schwartz JN et al (2007) American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43PubMed Wolff AC, Hammond ME, Schwartz JN et al (2007) American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer. Arch Pathol Lab Med 131:18–43PubMed
46.
Zurück zum Zitat Liu CL, Montgomery KD, Natkunam Y et al (2005) TMA-Combiner, a simple software tool to permit analysis of replicate cores on tissue microarrays. Mod Pathol 18:1641–1648PubMed Liu CL, Montgomery KD, Natkunam Y et al (2005) TMA-Combiner, a simple software tool to permit analysis of replicate cores on tissue microarrays. Mod Pathol 18:1641–1648PubMed
47.
Zurück zum Zitat Nielsen TO, Hsu FD, Jensen K et al (2004) Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10:5367–5374PubMedCrossRef Nielsen TO, Hsu FD, Jensen K et al (2004) Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res 10:5367–5374PubMedCrossRef
48.
Zurück zum Zitat Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea—a paradigm shift. Cancer Res 66:1883–1890 discussion 1895-6PubMedCrossRef Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea—a paradigm shift. Cancer Res 66:1883–1890 discussion 1895-6PubMedCrossRef
49.
Zurück zum Zitat Guler G, Balci S, Costinean S et al (2012) Stem cell-related markers in primary breast cancers and associated metastatic lesions. Mod Pathol 25:949–955PubMedCrossRef Guler G, Balci S, Costinean S et al (2012) Stem cell-related markers in primary breast cancers and associated metastatic lesions. Mod Pathol 25:949–955PubMedCrossRef
50.
Zurück zum Zitat Honeth G, Bendahl PO, Ringner M et al (2008) The CD44+/CD24− phenotype is enriched in basal-like breast tumors. Breast Cancer Res 10:R53PubMedCrossRef Honeth G, Bendahl PO, Ringner M et al (2008) The CD44+/CD24− phenotype is enriched in basal-like breast tumors. Breast Cancer Res 10:R53PubMedCrossRef
51.
Zurück zum Zitat Abraham BK, Fritz P, McClellan M et al (2005) Prevalence of CD44+/CD24−/low cells in breast cancer may not be associated with clinical outcome but may favor distant metastasis. Clin Cancer Res 11:1154–1159PubMed Abraham BK, Fritz P, McClellan M et al (2005) Prevalence of CD44+/CD24−/low cells in breast cancer may not be associated with clinical outcome but may favor distant metastasis. Clin Cancer Res 11:1154–1159PubMed
52.
Zurück zum Zitat Park SY, Lee HE, Li H et al (2010) Heterogeneity for stem cell-related markers according to tumor subtype and histologic stage in breast cancer. Clin Cancer Res 16:876–887PubMedCrossRef Park SY, Lee HE, Li H et al (2010) Heterogeneity for stem cell-related markers according to tumor subtype and histologic stage in breast cancer. Clin Cancer Res 16:876–887PubMedCrossRef
53.
Zurück zum Zitat Giatromanolaki A, Sivridis E, Fiska A et al (2011) The CD44+/CD24− phenotype relates to ‘triple-negative’ state and unfavorable prognosis in breast cancer patients. Med Oncol 28:745–752PubMedCrossRef Giatromanolaki A, Sivridis E, Fiska A et al (2011) The CD44+/CD24− phenotype relates to ‘triple-negative’ state and unfavorable prognosis in breast cancer patients. Med Oncol 28:745–752PubMedCrossRef
54.
Zurück zum Zitat Ricardo S, Vieira AF, Gerhard R et al (2011) Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol 64:937–946PubMedCrossRef Ricardo S, Vieira AF, Gerhard R et al (2011) Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol 64:937–946PubMedCrossRef
55.
Zurück zum Zitat Mylona E, Giannopoulou I, Fasomytakis E et al (2008) The clinicopathologic and prognostic significance of CD44+/CD24(−/low) and CD44−/CD24+ tumor cells in invasive breast carcinomas. Hum Pathol 39:1096–1102PubMedCrossRef Mylona E, Giannopoulou I, Fasomytakis E et al (2008) The clinicopathologic and prognostic significance of CD44+/CD24(−/low) and CD44−/CD24+ tumor cells in invasive breast carcinomas. Hum Pathol 39:1096–1102PubMedCrossRef
56.
Zurück zum Zitat Heerma van Voss MR, van der Groep P, Bart J et al (2011) Expression of the stem cell marker ALDH1 in BRCA1 related breast cancer. Cell Oncol (Dordr) 34:3–10CrossRef Heerma van Voss MR, van der Groep P, Bart J et al (2011) Expression of the stem cell marker ALDH1 in BRCA1 related breast cancer. Cell Oncol (Dordr) 34:3–10CrossRef
57.
Zurück zum Zitat Charafe-Jauffret E, Ginestier C, Iovino F et al (2010) Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 16:45–55PubMedCrossRef Charafe-Jauffret E, Ginestier C, Iovino F et al (2010) Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 16:45–55PubMedCrossRef
58.
Zurück zum Zitat Morimoto K, Kim SJ, Tanei T et al (2009) Stem cell marker aldehyde dehydrogenase 1-positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression. Cancer Sci 100:1062–1068PubMedCrossRef Morimoto K, Kim SJ, Tanei T et al (2009) Stem cell marker aldehyde dehydrogenase 1-positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression. Cancer Sci 100:1062–1068PubMedCrossRef
59.
Zurück zum Zitat Nalwoga H, Arnes JB, Wabinga H et al (2010) Expression of aldehyde dehydrogenase 1 (ALDH1) is associated with basal-like markers and features of aggressive tumours in African breast cancer. Br J Cancer 102:369–375PubMedCrossRef Nalwoga H, Arnes JB, Wabinga H et al (2010) Expression of aldehyde dehydrogenase 1 (ALDH1) is associated with basal-like markers and features of aggressive tumours in African breast cancer. Br J Cancer 102:369–375PubMedCrossRef
60.
Zurück zum Zitat Madjd Z, Ramezani B, Molanae S et al (2012) High expression of stem cell marker ALDH1 is associated with reduced BRCA1 in invasive breast carcinomas. Asian Pac J Cancer Prev 13:2973–2978PubMedCrossRef Madjd Z, Ramezani B, Molanae S et al (2012) High expression of stem cell marker ALDH1 is associated with reduced BRCA1 in invasive breast carcinomas. Asian Pac J Cancer Prev 13:2973–2978PubMedCrossRef
61.
Zurück zum Zitat Resetkova E, Reis-Filho JS, Jain RK et al (2010) Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat 123:97–108PubMedCrossRef Resetkova E, Reis-Filho JS, Jain RK et al (2010) Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat 123:97–108PubMedCrossRef
62.
Zurück zum Zitat Wright MH, Calcagno AM, Salcido CD et al (2008) Brca1 breast tumors contain distinct CD44+/CD24− and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10:R10PubMedCrossRef Wright MH, Calcagno AM, Salcido CD et al (2008) Brca1 breast tumors contain distinct CD44+/CD24− and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res 10:R10PubMedCrossRef
63.
Zurück zum Zitat Boecker W, Buerger H (2003) Evidence of progenitor cells of glandular and myoepithelial cell lineages in the human adult female breast epithelium: a new progenitor (adult stem) cell concept. Cell Prolif 36(Suppl 1):73–84PubMedCrossRef Boecker W, Buerger H (2003) Evidence of progenitor cells of glandular and myoepithelial cell lineages in the human adult female breast epithelium: a new progenitor (adult stem) cell concept. Cell Prolif 36(Suppl 1):73–84PubMedCrossRef
64.
Zurück zum Zitat Lim E, Vaillant F, Wu D et al (2009) Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med 15:907–913PubMedCrossRef Lim E, Vaillant F, Wu D et al (2009) Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat Med 15:907–913PubMedCrossRef
65.
Zurück zum Zitat Slamon DJ, Godolphin W, Jones LA et al (1989) Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244:707–712PubMedCrossRef Slamon DJ, Godolphin W, Jones LA et al (1989) Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 244:707–712PubMedCrossRef
66.
Zurück zum Zitat Goldhirsch A, Wood WC, Gelber RD et al (2007) Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer 2007. Ann Oncol 18:1133–1144PubMedCrossRef Goldhirsch A, Wood WC, Gelber RD et al (2007) Progress and promise: highlights of the international expert consensus on the primary therapy of early breast cancer 2007. Ann Oncol 18:1133–1144PubMedCrossRef
67.
Zurück zum Zitat Neumeister V, Agarwal S, Bordeaux J et al (2010) In situ identification of putative cancer stem cells by multiplexing ALDH1, CD44, and cytokeratin identifies breast cancer patients with poor prognosis. Am J Pathol 176:2131–2138PubMedCrossRef Neumeister V, Agarwal S, Bordeaux J et al (2010) In situ identification of putative cancer stem cells by multiplexing ALDH1, CD44, and cytokeratin identifies breast cancer patients with poor prognosis. Am J Pathol 176:2131–2138PubMedCrossRef
68.
Zurück zum Zitat Ridolfi RL, Rosen PP, Port A et al (1977) Medullary carcinoma of the breast: a clinicopathologic study with 10 year follow-up. Cancer 40:1365–1385PubMedCrossRef Ridolfi RL, Rosen PP, Port A et al (1977) Medullary carcinoma of the breast: a clinicopathologic study with 10 year follow-up. Cancer 40:1365–1385PubMedCrossRef
69.
Zurück zum Zitat Marginean F, Rakha EA, Ho BC et al (2010) Histological features of medullary carcinoma and prognosis in triple-negative basal-like carcinomas of the breast. Mod Pathol 23:1357–1363PubMedCrossRef Marginean F, Rakha EA, Ho BC et al (2010) Histological features of medullary carcinoma and prognosis in triple-negative basal-like carcinomas of the breast. Mod Pathol 23:1357–1363PubMedCrossRef
70.
Zurück zum Zitat Quenneville LA, Phillips KA, Ozcelik H et al (2002) HER-2/neu status and tumor morphology of invasive breast carcinomas in Ashkenazi women with known BRCA1 mutation status in the Ontario Familial Breast Cancer Registry. Cancer 95:2068–2075PubMedCrossRef Quenneville LA, Phillips KA, Ozcelik H et al (2002) HER-2/neu status and tumor morphology of invasive breast carcinomas in Ashkenazi women with known BRCA1 mutation status in the Ontario Familial Breast Cancer Registry. Cancer 95:2068–2075PubMedCrossRef
71.
Zurück zum Zitat Lakhani SR, Van De Vijver MJ, Jacquemier J et al (2002) The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol 20:2310–2318PubMedCrossRef Lakhani SR, Van De Vijver MJ, Jacquemier J et al (2002) The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol 20:2310–2318PubMedCrossRef
72.
Zurück zum Zitat Liu S, Lachapelle J, Leung S et al (2012) CD8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast Cancer Res 14:R48PubMedCrossRef Liu S, Lachapelle J, Leung S et al (2012) CD8+ lymphocyte infiltration is an independent favorable prognostic indicator in basal-like breast cancer. Breast Cancer Res 14:R48PubMedCrossRef
73.
Zurück zum Zitat West NR, Milne K, Truong PT et al (2011) Tumor-infiltrating lymphocytes predict response to anthracycline-based chemotherapy in estrogen receptor-negative breast cancer. Breast Cancer Res 13:R126PubMedCrossRef West NR, Milne K, Truong PT et al (2011) Tumor-infiltrating lymphocytes predict response to anthracycline-based chemotherapy in estrogen receptor-negative breast cancer. Breast Cancer Res 13:R126PubMedCrossRef
74.
Zurück zum Zitat Rody A, Holtrich U, Pusztai L et al (2009) T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res 11:R15PubMedCrossRef Rody A, Holtrich U, Pusztai L et al (2009) T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res 11:R15PubMedCrossRef
75.
Zurück zum Zitat van de Rijn M, Perou CM, Tibshirani R et al (2002) Expression of cytokeratins 17 and 5 identifies a group of breast carcinomas with poor clinical outcome. Am J Pathol 161:1991–1996PubMedCrossRef van de Rijn M, Perou CM, Tibshirani R et al (2002) Expression of cytokeratins 17 and 5 identifies a group of breast carcinomas with poor clinical outcome. Am J Pathol 161:1991–1996PubMedCrossRef
76.
Zurück zum Zitat Mulligan AM, Pinnaduwage D, Bull SB et al (2008) Prognostic effect of basal-like breast cancers is time dependent: evidence from tissue microarray studies on a lymph node-negative cohort. Clin Cancer Res 14:4168–4174PubMedCrossRef Mulligan AM, Pinnaduwage D, Bull SB et al (2008) Prognostic effect of basal-like breast cancers is time dependent: evidence from tissue microarray studies on a lymph node-negative cohort. Clin Cancer Res 14:4168–4174PubMedCrossRef
77.
Zurück zum Zitat Dent R, Hanna WM, Trudeau M et al (2009) Time to disease recurrence in basal-type breast cancers: effects of tumor size and lymph node status. Cancer 115:4917–4923PubMedCrossRef Dent R, Hanna WM, Trudeau M et al (2009) Time to disease recurrence in basal-type breast cancers: effects of tumor size and lymph node status. Cancer 115:4917–4923PubMedCrossRef
78.
Zurück zum Zitat Blows FM, Driver KE, Schmidt MK et al (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:e1000279PubMedCrossRef Blows FM, Driver KE, Schmidt MK et al (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:e1000279PubMedCrossRef
79.
Zurück zum Zitat Kubista M, Rosner M, Kubista E et al (2002) Brca1 regulates in vitro differentiation of mammary epithelial cells. Oncogene 21:4747–4756PubMedCrossRef Kubista M, Rosner M, Kubista E et al (2002) Brca1 regulates in vitro differentiation of mammary epithelial cells. Oncogene 21:4747–4756PubMedCrossRef
80.
Zurück zum Zitat Furuta S, Jiang X, Gu B et al (2005) Depletion of BRCA1 impairs differentiation but enhances proliferation of mammary epithelial cells. Proc Natl Acad Sci U S A 102:9176–9181PubMedCrossRef Furuta S, Jiang X, Gu B et al (2005) Depletion of BRCA1 impairs differentiation but enhances proliferation of mammary epithelial cells. Proc Natl Acad Sci U S A 102:9176–9181PubMedCrossRef
81.
Zurück zum Zitat Liu S, Ginestier C, Charafe-Jauffret E et al (2008) BRCA1 regulates human mammary stem/progenitor cell fate. Proc Natl Acad Sci U S A 105:1680–1685PubMedCrossRef Liu S, Ginestier C, Charafe-Jauffret E et al (2008) BRCA1 regulates human mammary stem/progenitor cell fate. Proc Natl Acad Sci U S A 105:1680–1685PubMedCrossRef
82.
Zurück zum Zitat Molyneux G, Geyer FC, Magnay FA et al (2010) BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell 7:403–417PubMedCrossRef Molyneux G, Geyer FC, Magnay FA et al (2010) BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell 7:403–417PubMedCrossRef
83.
Zurück zum Zitat Turner NC, Reis-Filho JS, Russell AM et al (2007) BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 26:2126–2132PubMedCrossRef Turner NC, Reis-Filho JS, Russell AM et al (2007) BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene 26:2126–2132PubMedCrossRef
Metadaten
Titel
Clinical–pathologic significance of cancer stem cell marker expression in familial breast cancers
verfasst von
Anita Bane
Alicia Viloria-Petit
Dushanthi Pinnaduwage
Anna Marie Mulligan
Frances P. O’Malley
Irene L. Andrulis
Publikationsdatum
01.07.2013
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 1/2013
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-013-2591-1

Weitere Artikel der Ausgabe 1/2013

Breast Cancer Research and Treatment 1/2013 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.