Skip to main content
Erschienen in: BMC Cancer 1/2017

Open Access 01.12.2017 | Research article

Prognostic significance of TRAIL-R3 and CCR-2 expression in tumor epithelial cells of patients with early breast cancer

verfasst von: Vivian Labovsky, Leandro Marcelo Martinez, Kevin Mauro Davies, María de Luján Calcagno, Hernán García-Rivello, Alejandra Wernicke, Leonardo Feldman, Ayelén Matas, María Belén Giorello, Francisco Raúl Borzone, Hosoon Choi, Scott C. Howard, Norma Alejandra Chasseing

Erschienen in: BMC Cancer | Ausgabe 1/2017

Abstract

Background

Tumor epithelial cells (TEpCs) and spindle-shaped stromal cells, not associated with the vasculature, of patients with early breast cancer express osteoprotegerin (OPG), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), receptor activator of nuclear factor kappa B ligand, stromal cell derived factor-1, interleukin-6, macrophage colony stimulating factor, chemokine (C-C motif) ligand-2 (CCL-2) and their receptors at significantly higher levels compared with non-neoplastic breast tissues. We evaluated the clinicopathological significance of these ligands and receptors in TEpC and spindle-shaped stromal cells, not associated with the vasculature, to determine their impact on prognosis of patients with early-stage breast cancer.

Methods

We conducted immunohistochemical analyses of protein expression in primary tumors of patients with early breast cancer and analyzed their association with standard prognostic parameters and clinical outcomes, including local relapse, metastatic recurrence, disease-free survival (DFS), metastasis-free survival (MFS), and overall survival (OS).

Results

Elevated levels of TRAIL-R3 and chemokine (C-C motif) receptor 2 (CCR-2) in TEpCs and OPG and CCL-2 in stromal cells were significantly associated with a higher risk of metastasis (p = 0.032, p = 0.003, p = 0.038, and p = 0.049; respectively). Moreover, high expression of TRAIL-R3 and CCR-2 in TEpCs was associated with shorter DFS, MFS, and OS. High TRAIL-R3 expression in TEpCs was an independent prognostic factor for DFS and OS, and high CCR-2 expression in these cells was an independent prognostic factor for MFS.

Conclusions

High levels of TRAIL-R3 and CCR-2 expression in TEpCs identified patients with early breast cancer with poor outcomes.
Abkürzungen
DFS
Disease-free survival
MFS
Metastasis-free survival
OS
Overall survival
TEpCs
Tumor epithelial cells

Background

Breast cancer is the most common cancer among women worldwide [14] and in Argentina affects more than 25,000 women and causes more than 5000 deaths each year (Bureau of Health Information Statistics and Nation, Department of Statistics and Health Information, Ministry of Health, Argentina, 2013). Distant metastasis is the main cause of death in these patients [5]. In high-income countries, breast cancer is usually diagnosed early, and treatment with curative intent and manageable toxicity is feasible. However, many women experience recurrence despite receiving optimal therapy, likely because the tumor microenvironment plays a key role in the development of resistance to treatment [6].
Breast cancer tissue comprises tumor epithelial cells (TEpCs) and stromal cells such as mesenchymal stem cells, tumor-associated fibroblasts, fibroblasts, endothelial cells, adipocytes, and immune cells. The interaction of malignant and non-malignant cells influences tumorigenesis, tumor growth, metastasis, and response to therapy [615]. Our group demonstrated that spindle-shaped stromal cells are not associated with the vasculature and TEpCs from primary invasive ductal breast cancer in women with stage I or II express molecules such as osteoprotegerin (OPG), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), receptor activator of nuclear factor kappa B ligand (RANKL), stromal cell derived factor (SDF)-1, interleukin (IL)-6, chemokine (C-C motif) ligand-2 (CCL-2) and their receptors [15]. These molecules, which are likely involved in the interactions between these cell types, mediate proliferation, survival, migration, and intravasation of TEpCs as well as angiogenesis in the primary tumor [15]. These findings led us to ask whether the levels of expression of these ligand-receptor pairs are useful for predicting the outcomes of patients with stage I/II breast cancer.

Methods

Patients

We conducted a retrospective study of 63 consecutive patients (aged 42–80 years) with a confirmed histological diagnosis of breast cancer who underwent initial surgery at the Hospital Italiano of Buenos Aires, Argentina. Patients were included if they were diagnosed with stage I/II invasive ductal breast cancer according to the International Union Against Cancer TNM classification system [16] and ≥10 years after surgery. Exclusion criteria included neoadjuvant therapies, lack of tissue, and another primary tumor. After surgery, all patients were treated with the indicated therapy, depending on their clinical status and the histopathological characteristics of their tumor, which were determined according to the recommendations of the European Society for Medical Oncology [17]. The Instituto de Biología y Medicina Experimental and the Hospital Italiano Ethics Committees approved this study, and informed consent was obtained from patients or the relatives of deceased patients, in accordance with the principles of the Helsinki Declaration. Physicians who were unaware of the pathology results acquired clinical information from patients’ medical records, and the anonymity of the data was ensured using a code made available only to the biostatistician.

Tumor samples

Breast tissues embedded in paraffin blocks and fixed in 10% neutral-buffered formalin were retrieved from the surgical archives, and 4-μm thick sections were used in the experiments described below.

Analysis of protein expression

These tissues were processed and immunohistochemistry was used to determine the levels of ligands and receptors in TEpC and in spindle-shaped stromal cells, not associated with the vasculature, and it was completed as described in a previously work [15].
Immunoreactivity was reviewed and scored independently by two pathologists who were blinded to patient outcomes. In uncertain cases, re-evaluation was performed using a double-headed microscope, and staining was evaluated until a consensus was achieved. The agreement in immunohistochemical evaluation between the two observers was 91.77% (Cohen’s kappa coefficient = 0.895). Each sample was assayed in duplicate and was initially examined at 100× magnification followed by observation of five representative fields at 400× magnification along a projected Z-line. Expression levels were evaluated separately for the TEpCs and spindle-shaped stromal cells, not associated to the vasculature, per the percentage of positive cells and staining intensity, which were estimated according to the Allred score [15, 16]. The percentages of positive cells were assigned scores as follows: 0 (<10%), 1 (10%–30%), 2 (31%–60%), 3 (61%–90%), and 4 (>90%). Staining intensity was scored as 0 (no staining), 1 (weak), 2 (moderate), and 3 (strong), according to the relative intensity of staining of TEpCs analyzed using the anti-cytokeratin antibody [15, 18]. The final staining score was calculated using the sum of the percentage of positive cells and the staining intensity score, which ranged from 0 to 7. Stromal cells included in this study had a spindle shape and were not associated with vasculature. CD34 expression was undetectable in this type of stromal cells as previously reported [18].

Patients’ clinicopathological characteristics

Classical prognostic markers were categorized according to cut-offs used in the protocols of the Hospital Italiano, [17] including: a) age < 50 or ≥50 years; b) tumor size <2 or ≥2 cm; c) histological grade according to the Scarff-Bloom-Richardson grading system [19], which is expressed as differentiated (G1), intermediate (G2), and poor (G3); d) expression of estrogen/progesterone receptors and HER2/neu was defined as negative or positive according to Wernicke et al. [17]; and e) presence of regional metastatic lymph nodes was recorded as negative (negative nodes in axillary dissection or sentinel lymph node) or positive (including micrometastasis) (Table 1).
Table 1
Clinical characteristics of 63 patients with early invasive ductal breast cancer
Characteristics
Patients (n)
Patients (%)
Age (years)
 < 50
10
15.9
 ≥ 50
53
84.1
 Unknown
-
-
Tumor size (cm)
 <2
45
71.4
 ≥ 2
17
27.0
 Unknown
1
1.6
Histological grade
 G1
15
23.8
 G2
22
34.9
 G3
24
38.1
 Unknown
2
3.2
HER2/neu status
 Negative
39
61.9
 Positive
23
36.5
 Unknown
1
1.6
ER status
 Negative
15
23.8
 Positive
47
74.6
 Unknown
1
1.6
PR status
 Negative
14
22.2
 Positive
48
76.2
 Unknown
1
1.6
Regional lymph nodes
 Negative
44
69.8
 Positive
16
25.4
 Unknown
3
4.8
Local relapse
 Negative
50
79.4
 Positive
6
9.5
 Unknown
7
11.1
Metastatic event
 Non-metastasis
45
71.4
 Metastasis
11
17.5
 Unknown
7
11.1

Statistical analysis

To evaluate the statistical significance of the associations between the expression of ligand or receptor and patients’ clinicopathological characteristics, we determined an optimal cut-off value according to a previous study [18]. The cut-off value was used to assign protein expression in tumor samples as negative/low or high. To determine the optimal cut-off value, the first quartile (Q1), median, and third quartile (Q3) values were tested individually using univariate analysis and compared with OS. The cut-off value with lowest p value was chosen. The optimal cut-off values for protein expression in TEpCs were as follows: OPG = 6 (Q3), TRAIL =6 (Q3), TRAIL-R1 = 0 (Q1), TRAIL-R2 = 6 (Q3), TRAIL-R3 = 5 (median), TRAIL-R4 = 5 (Q1), RANKL =3 (Q1), RANK =6 (Q1), SDF-1 = 5 (Q1), CXCR-4 = 4 (Q1), IL-6 = 4 (median), IL-6R = 6 (median), CCL-2 = 6 (Q3), and chemokine (C-C motif) receptor 2 (CCR-2) = 6 (Q3). The optimal cut-off values for protein expression in spindle-shaped stromal cells, not associated with vasculature, were as follows: OPG = 2 (median), TRAIL =4 (median), RANKL =2 (Q1), SDF-1 = 2 (Q1), IL-6 = 4 (Q3), and CCL-2 = 3 (Q3). We used Fisher’s exact test to evaluate the association between the expression of these proteins with classical prognostic markers as well as local relapse and metastatic occurrence. Moreover, the association between the ligand and receptor expressions in TEpCs and spindle-shaped stromal cells and metastatic occurrence is displayed as a heat map prepared using Excel.
DFS and MFS were defined as the interval from date of surgery to the first observation of tumor occurrence (metastatic occurrence and/or local relapse) and metastatic occurrence, respectively, or last follow-up. The interval from the date of surgery until death or last follow-up was defined as OS. Univariate analyses of DFS, MFS, and OS were performed using the Kaplan-Meier method, and the differences were evaluated using the log-rank (Mantel-Cox) test. When significant variables were identified, we applied the Cox proportional hazards model to the multivariate survival analysis using backward stepwise selection (likelihood ratio) that incorporated only the significant variables. Statistical analysis was performed using SSPS software (version 18.00, Chicago, Illinois) and InfoStat (version 2012, InfoStat Group, National University of Cordoba, Argentina). A two-sided p value <0.05 was considered statistically significant.

Results

Association of expression in TEpCs of OPG, TRAIL, RANKL, SDF-1, IL-6, and CCL-2 with patients’ clinicopathological characteristics

The expression of TRAIL was significantly associated with lymph node status (Table 2). High TRAIL expression was detected in 10/40 breast cancer patients with negative lymph nodes, and TRAIL expression was undetectable in 0/16 of patients with positive lymph nodes. SDF-1 expression was significantly associated with tumor size and was high in 31/44 patients with tumors <2 cm and in 7/17 patients with tumors ≥2 cm (p = 0.004, Table 2). High levels of CCL-2 expression were detected in some patients with negative (3/15) or positive (1/42) ER expression (Table 2). The DFS of patients with high CCL-2 expression was 67.7 ± 32.0 months, compared with 123.15 ± 8.28 months for those with low/negative CCL-2 expression (p = 0.048, Table 3).
Table 2
Association of the levels of expression of OPG, TRAIL, RANKL, SDF-1, IL-6, and CCL-2 in TEpCs with the clinicopathological characteristics of patients with early invasive
Characteristics
Ligands in TEpC
OPG
TRAIL
RANKL
SDF-1
IL-6
CCL-2
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
Age (years)
<50
10.0
>0.999
2.2
0.657
70.0
>0.999
40.0
0.153
22.2
0.464
22.2
0.110
≥50
15.7
16.0
73.1
67.3
39.2
4.1
Tumor size (cm)
<2
13.9
>0.999
17.1
>0.999
65.9
0.114
70.4
0.044*
42.9
0.236
7.3
>0.999
≥2
11.8
17.6
88.2
41.2
23.5
6.2
Histological grade
G1
20.0
0.713
21.4
0.744
46.7
0.073
80.0
0.093
42.9
0.307
6.7
0.357
G2
10.0
20.0
80.9
66.7
45.0
0.0
G3
12.5
13.0
79.2
45.8
25.0
13.0
HER2/neu status
Negative
8.1
0.239
18.9
0.733
76.3
0.388
63.2
>0.999
39.5
0.780
2.9
0.287
Positive
21.7
14.3
65.2
60.9
33.33
13.6
ER status
Negative
13.3
>0.999
7.7
0.429
80.0
0.523
40.0
0.064
42.9
0.753
20.0
0.049*
Positive
13.3
20.0
69.6
69.6
35.6
2.4
PR status
Negative
14.3
>0.999
8.3
0.444
78.6
0.737
50.0
0.351
38.5
>0.999
7.1
>0.999
Positive
13.0
19.6
70.2
66.0
37.0
7.0
Regional lymph nodes
Negative
11.6
0.666
25.0
0.048*
67.4
0.352
58.1
0.555
39.0
0.761
4.7
0.349
Positive
20.0
0.0
81.2
68.7
31.2
14.3
Metastatic occurrence
Negative
20.9
0.180
21.4
0.664
70.4
0.707
59.1
>0.999
30.2
>0.999
2.4
0.109
Positive
18.2
10.0
81.8
63.6
40.0
18.2
Local relapse
Negative
6.1
0.330
20.8
0.576
70.0
0.307
60.0
>0.999
38.8
0.284
4.3
0.266
Positive
20.0
0.0
100.0
60.0
0.0
20.0
Table 3
Univariate analysis of disease-free, metastasis-free, and overall survival of patients with early invasive ductal breast cancer
Univariate
p-value
Disease-free survival
Metastasis-free survival
Overall survival
Age
0.598
0.448
0.500
Tumor size
0.113
0.020*
0.069
Histological grade
0.178
0.291
0.207
HER2/neu status
0.536
0.293
0.103
ER status
0.336
0.191
0.175
PR status
0.691
0.946
0.521
Regional lymph nodes
0.595
0.805
0.620
OPG/TEpC
0.167
0.052
0.178
TRAIL/TEpC
0.465
0.648
0.304
RANKL/TEpC
0.156
0.267
0.307
SDF-1/TEpC
0.660
0.932
0.710
IL-6/TEpC CCL-2
0.873
0.710
0.487
CCL-2/TEpC
0.048*
0.071
0.507
TRAIL-R1/TEpC
0.536
0.339
0.626
TRAIL-R2/TEpC
0.894
0.749
0.392
TRAIL-R3/TEpC
0.009*
0.012*
0.015*
TRAIL-R4/TEpC
0.186
0.131
0.478
RANK/TEpC
0.546
0.991
0.804
CXCR-4/TEpC
0.164
0.255
0.175
IL-6R/TEpC
0.391
0.540
0.626
CCR-2/TEpC
0.013*
0.002*
0.049*
OPG/stromal cells
0.318
0.101
0.441
TRAIL/stromal cells
0.284
0.084
0.337
RANKL/stromal cells
0.139
0.052
0.222
SDF-1/stromal cells
0.792
0.734
0.306
IL-6/stromal cells CCL-2
0.218
0.093
0.168
CCL-2/stromal cells
0.104
0.076
0.505

Association of expression in TEpCs of TRAIL-R1–4, RANK, CXCR-4, IL-6R, and CCR-2 with patients’ clinicopathological characteristics

IL-6R expression in TEpCs was associated with age (Table 4). Specifically, IL-6R expression was higher in 6/8 patients <50 years of age and in 15/48 patients ≥50 years of age (Table 4). Patients with high expression of TRAIL-R3 and CCR-2 in TEpCs were at significantly higher risk for metastatic tumors than patients with low expression (Table 4). High levels of TRAIL-R3 were expressed in 7/11 breast cancer patients with metastasis and in 12/45 patients with non-metastatic tumors (p = 0.032, Table 4). Certain patients with metastatic (5/11) or non-metastatic tumors (2/42) expressed high levels of CCR-2 (Fig. 1 and Table 4). There was an association of high TRAIL-R3 expression with shorter DFS, MFS, and OS (Table 3). The values of DFS, MFS and OS of patients with high TRAIL-R3 expression were as follows (months): 90.04 ± 14.64, 97.02 ± 14.08 and 112.75 ± 12.73, respectively; for patients with low/negative expression were 136.22 ± 7.52, 140.22 ± 6.61 and 146.51 ± 5.16, respectively (Fig. 2 and Table 3).
Table 4
Association of the levels of expression of TRAIL-R1-R4, RANK, CXCR-4, IL-6R, and CCR-2 in TEpCs with the clinicopathological characteristics of patients with early invasive ductal breast cancer
Characteristics
Receptors in TEpC
TRAIL-R1
TRAIL-R2
TRAIL-R3
TRAIL-R4
RANK
CXCR-4
IL-6-R
CCR-2
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
Age (years)
<50
87.5
0.249
0.0
>0.999
40.0
0.729
80.0
0.175
30.0
0.176
50.0
0.259
75.0
0.042*
11.1
>0.999
≥50
63.5
9.6
34.0
54.7
55.8
73.1
31.3
12.0
Tumor size (cm)
<2
66.7
>0.999
9.3
>0.999
35.6
>0.999
57.8
>0.999
50.0
0.579
68.2
>0.999
31.6
0.365
11.9
>0.999
≥2
64.7
5.9
35.3
58.8
58.8
70.6
47.1
12.5
Histological grade
G1
71.4
0.397
13.3
0.245
33.3
>0.999
60.0
0.803
53.3
>0.999
66.7
0.941
38.5
0.078
20.0
0.597
G2
55.0
0.0
36.7
63.6
52.4
71.4
16.7
10.0
G3
75.0
12.5
37.5
54.2
50.0
66.7
50.0
8.7
HER2/neu status
Negative
59.5
0.255
5.4
0.361
25.6
0.054
51.3
0.190
52.6
>0.999
71.0
0.776
26.5
0.083
16.7
0.235
Positive
77.3
13.0
52.2
69.6
52.2
65.2
52.4
4.5
ER status
Negative
66.7
>0.999
13.3
0.590
53.3
0.125
60.0
>0.999
60.0
0.562
73.3
0.757
46.7
0.361
20.0
0.359
Positive
65.9
6.7
29.8
57.4
50.0
67.4
32.5
9.3
PR status
Negative
64.3
>0.999
14.3
0.581
42.9
0.539
57.1
>0.999
57.1
0.766
78.6
0.516
35.7
>0.999
14.3
>0.999
Positive
66.7
6.5
33.3
58.3
51.1
66.0
36.6
11.4
Regional lymph nodes
Negative
64.3
0.342
9.3
>0.999
36.4
>0.999
61.4
0.771
55.8
0.558
72.1
0.533
35.0
0.749
11.9
>0.999
Positive
80.0
6.7
37.5
56.2
43.7
62.5
42.9
13.3
Metastatic occurrence
Negative
66.7
0.108
4.6
0.502
26.7
0.032*
51.1
0.092
50.0
>0.999
70.4
0.473
30.8
0.171
4.8
0.003*
Positive
90.9
9.1
63.6
81.8
45.4
54.5
54.5
45.4
Local relapse
Negative
70.8
>0.999
4.1
0.257
31.4
0.323
56.9
>0.999
48.0
0.669
70.0
0.316
35.6
>0.999
14.6
0.601
Positive
80.0
20.0
60.0
60.0
60.0
40.0
40.0
0.0
Furthermore, there was an association of high CCR-2 expression with shorter DFS, MFS and OS (Table 3). The values of DFS, MFS, and OS of patients with high CCR-2 expression were as follows (months): 87.57 ± 18.57, 87.71 ± 18.58, and 114.67 ± 15.29, respectively; for patients with low/negative expression were 127.57 ± 8.42, 133.94 ± 7.52, and 140.44 ± 6.41, respectively (Fig. 3 and Table 3).

Association of expression in spindle-shaped stromal cells of OPG, TRAIL, RANKL, SDF-1, IL-6, and CCL-2 with patients’ clinicopathological characteristics

SDF-1 expression in spindle-shaped stromal cells was associated with histological grades, and high SDF-1 expression was detected in 10/15, 14/21, and 8/24 patients with differentiation grades G1, G2, and G3, respectively (Table 5). In contrast, high expression of OPG and CCL-2 in stromal cells was associated with a higher risk of metastasis (Fig. 1 and Table 5). High expression of OPG was observed in 7/10 patients with metastatic tumors and in 14/43 patients with non-metastatic tumors (p = 0.038, Fig. 1 and Table 5). In patients with metastatic or non-metastatic tumors, 4/11 and 4/44 expressed high levels of CCL-2, respectively (Fig. 1 and Table 5).
Table 5
Association of the levels of expression of OPG, TRAIL, RANKL, SDF-1, IL-6, and CCL-2 in spindle-shaped stroma cells (not associated with the vasculature) with the clinicopathological characteristics of patients with early invasive ductal breast cancer
Characteristics
Ligands in spindle-shaped stromal cells
OPG
TRAIL
RANKL
SDF-1
IL-6
CCL-2
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
High expression %
p
Age (years)
<50
50.0
0.742
66.7
0.137
70.0
0.709
40.0
0.493
22.2
0.664
22.2
0.646
≥50
44.0
35.3
75.0
55.8
17.6
16.3
Tumor size (cm)
<2
48.8
0.255
38.1
>0.999
68.2
0.193
56.8
0.391
21.4
0.483
19.5
0.708
≥2
31.2
41.2
88.2
41.2
11.8
12.5
Histological grade
G1
60.0
0.336
35.7
0.518
60.0
0.491
66.7
0.042*
21.4
0.741
33.3
0.116
G2
35.0
50.0
76.2
66.7
20.0
5.3
G3
39.1
33.3
79.2
33.3
12.5
17.4
HER2/neu status
Negative
44.4
>0.999
44.7
0.272
78.9
0.368
57.9
0.302
18.4
>0.999
17.1
>0.999
Positive
43.5
28.6
65.2
43.5
19.0
18.2
ER status
Negative
46.7
>0.999
42.9
0.761
80.0
0.738
53.3
>0.999
21.4
>0.999
20.0
>0.999
Positive
43.2
37.8
71.7
52.2
17.8
16.7
PR status
Negative
42.9
>0.999
30.8
0.540
78.6
0.742
50.0
>0.999
15.4
>0.999
7.1
0.422
Positive
44.4
41.3
72.3
53.2
19.6
20.9
Regional lymph nodes
Negative
46.1
0.547
41.5
>0.999
74.4
0.745
55.8
0.773
17.1
>0.999
19.0
0.724
Positive
35.7
37.5
68.7
50.0
18.7
14.3
Metastatic occurrence
Negative
32.5
0.038*
34.9
0.072
65.9
0.146
47.3
0.745
11.6
0.163
9.1
0.049*
Positive
70.0
70.0
90.9
54.5
30.0
36.4
Local relapse
Negative
39.6
>0.999
42.9
0.633
72.0
0.621
50.0
>0.999
16.3
0.612
14.9
>0.999
Positive
40.0
25.0
60.0
40.0
0.0
20.0

Univariate analysis of the association of classical prognostic markers with DFS, MFS, and OS

Of clinical variables analyzed, only tumor size was associated with MFS (Table 3). Patients with tumors >2 cm had earlier metastasis compared with those with tumors ≤2 cm as follows (months): 93.00 ± 15.59 vs 139.02 ± 6.47, respectively.

Multivariate analysis

TRAIL-R3 expression in TEpCs was an independent prognostic factor for DFS and OS (Table 6). Moreover, tumor size and CCR-2 expression were independent prognostic factors for MFS (Table 6).
Table 6
Multivariate analysis of DFS, MFS, and OS of patients with early invasive ductal breast cancer
 
Variables
HR
95% CI
p
Disease-free survival
TRAIL-R3 in TEpC
3.566
1.164–10.920
0.026
Metastasis-free survival
Tumor size
8.210
2.013–33.477
0.003
CCR-2 in TEpC
10.257
2.569–40.947
0.001
Overall survival
TRAIL-R3 in TEpC
5.741
1.113–29.621
0.037
C.I. confidence interval, HR hazard ratio

Discussion

Tumor progression is a multistep process involving interactions between tumor cells and spindle-shaped stromal cells, not associated with the vasculature, which supply signals that may promote tumor progression [15].
Here we show that high TRAIL expression in TEpCs was significantly associated with negative lymph-node status. Paracrine signaling induced by the binding of TRAIL to the death receptors TRAIL-R1 and TRAIL-R2 induces apoptosis [2022]. Thus, the association of TRAIL expression in TEpCs of patients with negative lymph nodes might reflect the apoptotic effects of TRAIL that delay tumor progression as well as the extravasation of tumor cells to regional lymph nodes [23].
Patients with TEpCs that expressed high levels of TRAIL-R3 harbored metastases and experienced shorter DFS, MFS, and OS. TRAIL-R3 competes with TRAIL-R1, TRAIL-R2, or both for the binding of TRAIL, which inhibits apoptotic signaling [20]. Moreover, the expression of TRAIL-R3 in TEpCs was an independent prognostic marker for DFS and OS. These findings indicate the importance of evaluating TRAIL-R3 expression in TEpCs, because TRAIL is used to treat tumors. Thus, outcomes may be adversely affected by the level of TRAIL-R3 activity in tumors as well as in the tumor microenvironment.
In contrast, we found that high SDF-1 expression in TEpCs was significantly associated with tumor size <2 cm, which is consistent with the findings of previous studies [24, 25]. Furthermore, high expression of SDF-1 in spindle-shaped stromal cells, not associated with the vasculature, was significantly associated with conventional prognostic markers of less adverse tumor phenotypes, such as low histological grade (G1 and G2).
We show here that the expression of CCL-2 in TEpCs was associated with negative ER-status, which agrees with reports demonstrating that CCL-2 is overexpressed in ER-negative compared with ER-positive tumors [26]. These data suggest the involvement of CCL-2 in the progression of ER-negative breast tumors. Moreover high CCL-2 expression in TEpCs was significantly associated with DFS. CCL-2 directly promotes the malignant phenotype (epithelial mesenchymal transition) of TEpCs and increases their ability to migrate, proliferate, and invade tissues [2730]. Also, patients with high CCR-2 expression in TEpCs experienced shorter DFS, MFS, and OS. Furthermore, the expression of CCR-2 is up-regulated in breast tumor cells, and knockdown of CCR-2 expression inhibits breast tumor development [31]. Additionally, we show here that CCR-2 expression was an independent prognostic factor for MFS.
Stromal cells such as fibroblast that produce CCL-2 enhance the invasiveness and metastatic growth of human breast cancer cell lines [31], which is consistent with the present findings of a significant association between high CCL-2 expressions in spindle-shaped stromal cells in patients with metastatic early-stage breast cancer. Our data indicate the importance of evaluating CCR-2 expression in TEpCs as well as CCL-2 expression in TEpC and spindle-shaped stromal cells, because the pathways that produce CCR-2 and its ligands may provide targets for the prevention of breast cancer progression and metastasis [29]. Interestingly, we found previously that the expression of CCL-2 in spindle-shaped stromal cells, not associated with the vasculature, correlated positively with the expression of CCR-2 in TEpCs, suggesting that CCL-2 signaling through CCR-2 may contribute to the interactions between TEpCs and spindle-shaped stromal cells, which enhance the malignant phenotype of tumor cells during the early stages of disease [15].
We uncovered a significant association between high OPG expression in spindle-shaped stromal cells and the presence of metastatic breast tumors. This finding is consistent with those showing that OPG produced by a breast tumor induces angiogenesis and inhibits TRAIL-mediated apoptosis to promote the growth of the primary tumor as well as metastatic cells [32, 33].
To our knowledge, this study is the first to demonstrate that high expression of TRAIL-R3 and CCR-2 in TEpCs serves as a prognostic marker of metastatic tumors as well as DFS, MFS, and OS in women with stage I/II invasive breast cancer. These new findings provide a rationale for further studies designed to target TRAIL-R3 and CCR-2 signaling pathways to facilitate the diagnosis, prevention, and treatment of breast cancer.

Conclusions

High levels of TRAIL-R3 and CCR-2 expression in TEpCs identified early breast cancer patients with poor outcomes, including a higher risk of metastasis and shorter DFS, MFS, and OS and represent new independent prognostic factors that may also be suitable therapeutic targets.

Acknowledgments

The authors have no acknowledgements.

Funding

This research was supported by grants from Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), PIP 166 (2011–2013), Argentina; Fundación Alberto J. Roemmers (2009–2011)/(2010–2012)/(2012–2014), Argentina and Fundación Florencio Fiorini (2013), Argentina; Fundación René Barón (2015–2017), Argentina.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

Authors’ contrib

VL contributed to the conception and design of the study, performed the immunoassays and statistical analysis, and helped to draft the manuscript. LMM participated in the design of the study, performed the statistical analysis, coordination and helped to draft the manuscript. MLC performed the statistical analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis). KMD acquired clinical data and evaluation of the samples. HGR acquired clinical data and evaluation of the samples. AW acquired clinical data and evaluation of the samples. LF coordinated the study and helped to draft the manuscript. AM carried out the immunoassays. MBG contributed to the statistical analysis. FRB helped to draft the manuscript and analyze the data. HC helped to draft the manuscript and analyze the data. HSC helped to draft the manuscript and analyze the data. NAC participated in the design, conception, and conduct of the study and helped to draft the manuscript. All authors read and approved the final version of the manuscript.

Competing interests

The authors declare that they have no conflict of interest.
Written informed consents were obtained from the patient for publication of this case series. A copy of the written consent is available for review by the Editor of this journal.
This study was approved by the ethics committee of Hospital Italiano and Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and conducted according to the Declaration of Helsinki.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94(2):153–6.CrossRefPubMed Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94(2):153–6.CrossRefPubMed
2.
Zurück zum Zitat Bhatia P, Sanders MM, Hansen MF. Expression of receptor activator of nuclear factor-kappaB is inversely correlated with metastatic phenotype in breast carcinoma. Clin Cancer Res. 2005;11(1):162–5.PubMed Bhatia P, Sanders MM, Hansen MF. Expression of receptor activator of nuclear factor-kappaB is inversely correlated with metastatic phenotype in breast carcinoma. Clin Cancer Res. 2005;11(1):162–5.PubMed
3.
Zurück zum Zitat Kakarala M, Wicha MS. Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy. J Clin Oncol. 2008;26(17):2813–20.CrossRefPubMedPubMedCentral Kakarala M, Wicha MS. Implications of the cancer stem-cell hypothesis for breast cancer prevention and therapy. J Clin Oncol. 2008;26(17):2813–20.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Patel SA, Heinrich AC, Reddy BY, Srinivas B, Heidaran N, Rameshwar P. Breast cancer biology: the multifaceted roles of mesenchymal stem cells. J Oncol. 2008;2008:425895.CrossRefPubMedPubMedCentral Patel SA, Heinrich AC, Reddy BY, Srinivas B, Heidaran N, Rameshwar P. Breast cancer biology: the multifaceted roles of mesenchymal stem cells. J Oncol. 2008;2008:425895.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Rosa Mendoza ES, Moreno E, Caguioa PB. Predictors of early distant metastasis in women with breast cancer. J Cancer Res Clin Oncol. 2013;139(4):645–52.CrossRefPubMed Rosa Mendoza ES, Moreno E, Caguioa PB. Predictors of early distant metastasis in women with breast cancer. J Cancer Res Clin Oncol. 2013;139(4):645–52.CrossRefPubMed
6.
Zurück zum Zitat Nantajit D, Lin D, Li JJ. The network of epithelial-mesenchymal transition: potential new targets for tumor resistance. J Cancer Res Clin Oncol. 2015;141(10):1697–713.CrossRefPubMed Nantajit D, Lin D, Li JJ. The network of epithelial-mesenchymal transition: potential new targets for tumor resistance. J Cancer Res Clin Oncol. 2015;141(10):1697–713.CrossRefPubMed
7.
Zurück zum Zitat Beacham DA, Cukierman E. Stromagenesis: the changing face of fibroblastic microenvironments during tumor progression. Semin Cancer Biol. 2005;15(5):329–41.CrossRefPubMed Beacham DA, Cukierman E. Stromagenesis: the changing face of fibroblastic microenvironments during tumor progression. Semin Cancer Biol. 2005;15(5):329–41.CrossRefPubMed
8.
9.
Zurück zum Zitat Reddy BY, Lim PK, Silverio K, Patel SA, Won BW, Rameshwar P. The microenvironmental effect in the progression, metastasis, and dormancy of breast cancer: a model system within bone marrow. Int J Breast Cancer. 2012;2012:721659.CrossRefPubMedPubMedCentral Reddy BY, Lim PK, Silverio K, Patel SA, Won BW, Rameshwar P. The microenvironmental effect in the progression, metastasis, and dormancy of breast cancer: a model system within bone marrow. Int J Breast Cancer. 2012;2012:721659.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS, Burow ME. Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. Mol Cancer. 2010;9:295.CrossRefPubMedPubMedCentral Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS, Burow ME. Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. Mol Cancer. 2010;9:295.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Senst C, Nazari-Shafti T, Kruger S, Höner Zu Bentrup K, Dupin CL, et al. Prospective dual role of mesenchymal stem cells in breast tumor microenvironment. Breast Cancer Res Treat. 2013;137(1):69–79.CrossRefPubMed Senst C, Nazari-Shafti T, Kruger S, Höner Zu Bentrup K, Dupin CL, et al. Prospective dual role of mesenchymal stem cells in breast tumor microenvironment. Breast Cancer Res Treat. 2013;137(1):69–79.CrossRefPubMed
12.
Zurück zum Zitat Peng Q, Zhao L, Hou Y, Sun Y, Wang L, Luo H, et al. Biological characteristics and genetic heterogeneity between carcinoma-associated fibroblasts and their paired normal fibroblasts in human breast cancer. PLoS One. 2013;8(4):e60321.CrossRefPubMedPubMedCentral Peng Q, Zhao L, Hou Y, Sun Y, Wang L, Luo H, et al. Biological characteristics and genetic heterogeneity between carcinoma-associated fibroblasts and their paired normal fibroblasts in human breast cancer. PLoS One. 2013;8(4):e60321.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Barcellos-de-Souza P, Gori V, Bambi F, Chiarugi P. Tumor microenvironment: bone marrow-mesenchymal stem cells as key players. Biochim Biophys Acta. 2013;1836(2):321–35.PubMed Barcellos-de-Souza P, Gori V, Bambi F, Chiarugi P. Tumor microenvironment: bone marrow-mesenchymal stem cells as key players. Biochim Biophys Acta. 2013;1836(2):321–35.PubMed
15.
Zurück zum Zitat Labovsky V, Martinez LM, Davies KM, García-Rivello H, Calcagno Mde L, Matas A, et al. Association between Ligands and receptors related to the progression of early breast cancer in tumor epithelial and Stromal cells. Clin Breast Cancer. 2015;15(1):e13–21.CrossRefPubMed Labovsky V, Martinez LM, Davies KM, García-Rivello H, Calcagno Mde L, Matas A, et al. Association between Ligands and receptors related to the progression of early breast cancer in tumor epithelial and Stromal cells. Clin Breast Cancer. 2015;15(1):e13–21.CrossRefPubMed
16.
Zurück zum Zitat Edge S, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC cancer staging manual and handbook, seventh edition, American joint committee on cancer. New York: Springer-Verlag; 2010. Edge S, Byrd DR, Compton CC, Fritz AG, Greene FL, Trotti A. AJCC cancer staging manual and handbook, seventh edition, American joint committee on cancer. New York: Springer-Verlag; 2010.
17.
Zurück zum Zitat Wernicke M, Roitman P, Manfre D, Stern R. Breast cancer and the stromal factor. The "prometastatic healing process" hypothesis. Medicina (B Aires). 2011;71(1):15-21. Wernicke M, Roitman P, Manfre D, Stern R. Breast cancer and the stromal factor. The "prometastatic healing process" hypothesis. Medicina (B Aires). 2011;71(1):15-21.
18.
Zurück zum Zitat Martinez LM, Labovsky V, Calcagno ML, Davies KM, Garcia Rivello H, Bianchi MS, et al. CD105 expression on CD34-negative spindle-shaped stromal cells of primary tumor is an unfavorable prognostic marker in early breast cancer patients. PLoS One. 2015;10(3):e0121421.CrossRefPubMedPubMedCentral Martinez LM, Labovsky V, Calcagno ML, Davies KM, Garcia Rivello H, Bianchi MS, et al. CD105 expression on CD34-negative spindle-shaped stromal cells of primary tumor is an unfavorable prognostic marker in early breast cancer patients. PLoS One. 2015;10(3):e0121421.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Bloom HJ, Richardson WW. Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer. 1957;11(3):359–77.CrossRefPubMedPubMedCentral Bloom HJ, Richardson WW. Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer. 1957;11(3):359–77.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Labovsky V, Vallone VB, Martinez LM, Otaegui J, Chasseing NA. Expression of osteoprotegerin, receptor activator of nuclear factor kappa-B ligand, tumor necrosis factor-related apoptosis-inducing ligand, stromal cell-derived factor-1 and their receptors in epithelial metastatic breast cancer cell lines. Cancer Cell Int. 2012;12(1):29.CrossRefPubMedPubMedCentral Labovsky V, Vallone VB, Martinez LM, Otaegui J, Chasseing NA. Expression of osteoprotegerin, receptor activator of nuclear factor kappa-B ligand, tumor necrosis factor-related apoptosis-inducing ligand, stromal cell-derived factor-1 and their receptors in epithelial metastatic breast cancer cell lines. Cancer Cell Int. 2012;12(1):29.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Herbst RS, Eckhardt SG, Kurzrock R, Ebbinghaus S, O'Dwyer PJ, Gordon MS, et al. Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. J Clin Oncol. 2010;28(17):2839–46.CrossRefPubMed Herbst RS, Eckhardt SG, Kurzrock R, Ebbinghaus S, O'Dwyer PJ, Gordon MS, et al. Phase I dose-escalation study of recombinant human Apo2L/TRAIL, a dual proapoptotic receptor agonist, in patients with advanced cancer. J Clin Oncol. 2010;28(17):2839–46.CrossRefPubMed
22.
Zurück zum Zitat Garimella SV, Gehlhaus K, Dine JL, et al. Identification of novel molecular regulators of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in breast cancer cells by RNAi screening. Breast Cancer Res. 2014;16:R41.CrossRefPubMedPubMedCentral Garimella SV, Gehlhaus K, Dine JL, et al. Identification of novel molecular regulators of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in breast cancer cells by RNAi screening. Breast Cancer Res. 2014;16:R41.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Cross SS, Harrison RF, Balasubramanian SP, Lippitt JM, Evans CA, Reed MW, et al. Expression of receptor activator of nuclear factor kappabeta ligand (RANKL) and tumour necrosis factor related, apoptosis inducing ligand (TRAIL) in breast cancer, and their relations with osteoprotegerin, oestrogen receptor, and clinicopathological variables. J Clin Pathol. 2006;59(7):716–20.CrossRefPubMedPubMedCentral Cross SS, Harrison RF, Balasubramanian SP, Lippitt JM, Evans CA, Reed MW, et al. Expression of receptor activator of nuclear factor kappabeta ligand (RANKL) and tumour necrosis factor related, apoptosis inducing ligand (TRAIL) in breast cancer, and their relations with osteoprotegerin, oestrogen receptor, and clinicopathological variables. J Clin Pathol. 2006;59(7):716–20.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Lv ZD, Kong B, Liu XP, Dong Q, Niu HT, Wang YH, et al. CXCL12 chemokine expression suppresses human breast cancer growth and metastasis in vitro and in vivo. Int J Clin Exp Pathol. 2014;7(10):6671–8.PubMedPubMedCentral Lv ZD, Kong B, Liu XP, Dong Q, Niu HT, Wang YH, et al. CXCL12 chemokine expression suppresses human breast cancer growth and metastasis in vitro and in vivo. Int J Clin Exp Pathol. 2014;7(10):6671–8.PubMedPubMedCentral
25.
Zurück zum Zitat Mirisola V, Zuccarino A, Bachmeier BE, Sormani MP, Falter J, Nerlich A, et al. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur J Cancer. 2009;45(14):2579–87.CrossRefPubMed Mirisola V, Zuccarino A, Bachmeier BE, Sormani MP, Falter J, Nerlich A, et al. CXCL12/SDF1 expression by breast cancers is an independent prognostic marker of disease-free and overall survival. Eur J Cancer. 2009;45(14):2579–87.CrossRefPubMed
26.
Zurück zum Zitat Chavey C, Bibeau F, Gourgou-Bourgade S, Burlinchon S, Boissière F, Laune D, et al. Oestrogen receptor negative breast cancers exhibit high cytokine content. Breast Cancer Res. 2007;9(1):R15.CrossRefPubMedPubMedCentral Chavey C, Bibeau F, Gourgou-Bourgade S, Burlinchon S, Boissière F, Laune D, et al. Oestrogen receptor negative breast cancers exhibit high cytokine content. Breast Cancer Res. 2007;9(1):R15.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008;267(2):271–85.CrossRefPubMed Soria G, Ben-Baruch A. The inflammatory chemokines CCL2 and CCL5 in breast cancer. Cancer Lett. 2008;267(2):271–85.CrossRefPubMed
28.
Zurück zum Zitat Potter SM, Dwyer RM, Hartmann MC, Khan S, Boyle MP, Curran CE, et al. Influence of stromal-epithelial interactions on breast cancer in vitro and in vivo. Breast Cancer Res Treat. 2012;131(2):401–11.CrossRefPubMed Potter SM, Dwyer RM, Hartmann MC, Khan S, Boyle MP, Curran CE, et al. Influence of stromal-epithelial interactions on breast cancer in vitro and in vivo. Breast Cancer Res Treat. 2012;131(2):401–11.CrossRefPubMed
29.
Zurück zum Zitat Lu X, Kang Y. Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem. 2009;284(42):29087–96.CrossRefPubMedPubMedCentral Lu X, Kang Y. Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone. J Biol Chem. 2009;284(42):29087–96.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, Meyn RE, et al. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res. 2007;67(24):11687–95.CrossRefPubMedPubMedCentral Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, Meyn RE, et al. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res. 2007;67(24):11687–95.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Fang WB, Jokar I, Zou A, Lambert D, Dendukuri P, Cheng N. CCL2/CCR2 chemokine signaling coordinates survival and motility of breast cancer cells through Smad3 protein- and p42/44 mitogen-activated protein kinase (MAPK)-dependent mechanisms. J Biol Chem. 2012;287(43):36593–608.CrossRefPubMedPubMedCentral Fang WB, Jokar I, Zou A, Lambert D, Dendukuri P, Cheng N. CCL2/CCR2 chemokine signaling coordinates survival and motility of breast cancer cells through Smad3 protein- and p42/44 mitogen-activated protein kinase (MAPK)-dependent mechanisms. J Biol Chem. 2012;287(43):36593–608.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Weichhaus M, Segaran P, Renaud A, Geerts D, Connelly L. Osteoprotegerin expression in triple-negative breast cancer cells promotes metastasis. Cancer Med. 2014;3(5):1112–25.CrossRefPubMedPubMedCentral Weichhaus M, Segaran P, Renaud A, Geerts D, Connelly L. Osteoprotegerin expression in triple-negative breast cancer cells promotes metastasis. Cancer Med. 2014;3(5):1112–25.CrossRefPubMedPubMedCentral
Metadaten
Titel
Prognostic significance of TRAIL-R3 and CCR-2 expression in tumor epithelial cells of patients with early breast cancer
verfasst von
Vivian Labovsky
Leandro Marcelo Martinez
Kevin Mauro Davies
María de Luján Calcagno
Hernán García-Rivello
Alejandra Wernicke
Leonardo Feldman
Ayelén Matas
María Belén Giorello
Francisco Raúl Borzone
Hosoon Choi
Scott C. Howard
Norma Alejandra Chasseing
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2017
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3259-8

Weitere Artikel der Ausgabe 1/2017

BMC Cancer 1/2017 Zur Ausgabe

Update Onkologie

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