Open Access

Prognostic role of matrix metalloproteinase 9 in early breast cancer

  • Authors:
    • Katarina Kalavska
    • Zuzana Cierna
    • Marian Karaba
    • Gabriel Minarik
    • Juraj Benca
    • Tatiana Sedlackova
    • Denisa Kolekova
    • Ivana Mrvova
    • Daniel Pindak
    • Jozef Mardiak
    • Michal Mego
  • View Affiliations

  • Published online on: November 30, 2020     https://doi.org/10.3892/ol.2020.12339
  • Article Number: 78
  • Copyright: © Kalavska et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MMP9 is involved in extracellular matrix degradation during various physiological and pathological conditions, including tumorigenesis. The present study aimed to assess the prognostic role of intratumoral MMP9 and to determine its association with circulating tumor cells (CTCs) in patients with early breast cancer. A total of 318 patients with primary breast cancer (PBC) were enrolled into the present study. Specimens were subjected to immunohistochemistry analysis, using the MMP9 monoclonal antibody. MMP9 expression was scored using a weighted histoscore (WH). The results demonstrated that the mean WH ± SEM for MMP9 expression was significantly higher in breast tumor cells compared with tumor associated stromas (132.0±5.2 vs. 50.8±3.7; P<0.00001). Furthermore, a positive association was observed between MMP9 expression, the hormone positive status and proliferation index of analysed breast cancer tumour cells. Notably, the prognostic role of MMP9 was not observed in tumor cells [hazard ratio (HR) =0.96; 95% confidence interval (CI), 0.58‑1.59; P=0.864] or tumor associated stroma (HR=1.29; 95% CI, 0.60‑2.78; P=0.547). Subgroup analysis demonstrated that patients that were HR negative or triple negative, with low MMP9 expression in tumor cells and stroma had a significantly improved disease‑free survival than patients with high MMP9 expression. Taken together, the results of the present study demonstrated that high MMP9 expression in PBC was associated with favorable tumor characteristics. However, the prognostic value of MMP9 was limited to only the HR negative and CTC epithelial‑to‑mesenchymal transition positive subgroups. Thus, analyzing MMP9 tumor expression may help identify patients with increased risk of disease recurrence in these subgroups.

Introduction

Breast cancer is the most common malignancy in women worldwide, whereby 2,088,849 new cases of invasive breast cancer and 626,679 mortalities were reported in 2018 (1,2). Tumor invasion and metastasis affect >90% of patients with breast cancer, and thus notably contribute to the high mortality rate (35). This metastatic disease remains incurable, and effective treatment for end-stage metastatic breast cancer are yet to be determined (68). The aggressiveness of a tumor is closely associated with its ability to evade natural barriers, to invade adjacent tissues and metastasize distant sites (9). The metastatic cascade is a multistep process where cancer cells escape from the primary tumor site to distant locations, where they can potentially establish new cancer colonies (10,11). Under optimal conditions, epithelial cancer cells detach from the primary tumor site, penetrate and migrate via peripheral circulation, and invade secondary sites, where they ultimately undergo extravasation and populate distant organs (11,12).

Proteolytic degradation of the basement membrane and extracellular matrix (ECM) is considered a crucial aspect of metastatic growth, which enables low anchorage of neoplastic cells (1317). Several cell-secreted proteolytic enzymes, including matrix metalloproteinases (MMPs) are implicated in the cleavage of ECM (13,18,19). Matrix metalloproteinase 9 (MMP9) is a member of the gelatinase subfamily of MMPs and is secreted by a variety of cell types in an inactive form that undergoes activation upon cleavage by different types of extracellular proteases (18,20). MMP9 activity is modulated via different biochemical molecules, including growth factors and cytokines (19,21). Notably, MMP9 is actively involved in the degradation of type IV collagen, which is a crucial component of the basement membrane (19,22). In addition, MMP9 facilitates the dissemination machinery, and is particularly involved in tumour invasion, tumour-induced angiogenesis, and immunomodulation of the tumour microenvironment, where it is implicated in the formation of so-called premetastatic niches (23,24). Previous studies have focused on the association between high MMP9 expression and the number of distant metastases in patients with breast cancer (2527), as well as poor prognosis (28,29). It has been speculated that circulating tumor cells (CTCs), which are responsible for distant metastasis formation, use MMPs to form new metastatic sites (19,30). In addition, a previous study demonstrated that elevated MMP1 expression is significantly associated with the presence of CTC_ epithelial-to-mesenchymal transition (EMT) cells in the peripheral blood of patients with primary breast cancer (PBC), as well as with poor prognostic features of their primary tumors (31). The present study aimed to assess MMP9 expression in tumor cells as well as tumor associated stroma of patients with PBC, and determine its association with the presence of CTCs in the peripheral blood of these patients and other clinicopathological characteristics. The prognostic value of MMP9 in patients with PBC was also assessed.

Patients and methods

Study patients

The present study (Protocol TRU-SK 002; Chair: Michal Mego) enrolled 318 patients with stages I–III PBC who underwent definitive surgery. The samples were collected from the National Cancer Institute (Bratislava, Slovakia) between March 2012 and February 2015. The paraphing embedded tumor tissue and CTCs status in peripheral blood were available for all patients included in the present study. Complete diagnostic evaluation was performed in all patients to exclude the presence of distant metastasis. Patients with concurrent malignancy in the last 5 years, other than non-melanoma skin cancer, were excluded from the present study. The clinicopathological data including age, tumor stage, histology, regional lymph node involvement, hormone receptor status and HER2 status were retrieved and tabulated from the patients' records after obtaining all the relevant ethical approvals. Breast cancer subtypes were identified by immunohistochemical staining (see below) and classified according to the ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up for early breast cancer (32).

The present study was reviewed and approved by the Institutional Review Board of the National Cancer Institute of Slovakia, Bratislava, Slovakia (TRUSK002, 20.6.2011). Written informed consent was provided by all patients prior to the study start.

Tumor pathology

Pathological review was performed at the Department of Patholoχgy, Faculty of Medicine, Comenius University, (Bratislava, Slovakia) by an experienced pathologist (ZC).

Tumor samples and tissue microarray construction

Tumor specimens used in the present study were classified according to the 2019 World Health Organization classification (33). According to the tumor histology results, one or two representative areas containing the most representative part of the hematoxylin and eosin (H&E) stained tumor tissues were observed under a light microscope, original magnification×400. The identified sections were matched to their corresponding wax blocks (donor blocks). The 3-mm diameter cores of the tumors were removed from the donor blocks using the multipurpose sampling tool HarrisUni-Core (Sigma-Aldrich; Merck KGaA) and inserted into the recipient master block. The recipient block was cut into 5-µm-thick sections, which were transferred onto coated slides.

Immunohistochemical (IHC) staining

Deparaffinized slides were rehydrated in phosphate buffered saline solution (10 mM, pH 7.2). Tissue epitopes were demasked using the automated water bath heating process in Dako PT Link (Dako; Agilent Technologies, Inc.) and the slides were incubated in pH 6.0 citrate retrieval buffer at 98°C for 20 min. The slides were subsequently incubated for 1 h at room temperature with primary mouse monoclonal antibody against MMP9 (Abcam; MMP9 (SB15c); cat. no. ab51203) diluted 1:200 in Dako REAL antibody diluent (Dako; Agilent Technologies, Inc.) and immunostained with anti-mouse/anti-rabbit immuno-peroxidase polymer (EnVision FLEX/HRP, Dako; Agilent Technologies, Inc.) for 30 min at room temperature, according to the manufacturer's instructions. The reaction was visualized using diaminobenzidine substrate-chromogen solution (Dako; Agilent Technologies, Inc.) for 5 min, and the slides were counterstained with hematoxylin. The human clone tissue served as the positive control, and colon tissue subjected to the same procedure omitting the primary antibody was used as the negative control.

IHC evaluation

Tumor scores were blindly assessed by a pathologist (ZC). The results of the IHC analyzes were scored using a weighted histoscore (WH), assessing both the percentage of positive cells (PP) and the staining intensity (SI) of the cytoplasm as follows: The proportion of cells with nuclear staining was multiplied by the intensity of staining to provide a histoscore ranging from 0–300. The histoscore was calculated as follows: Score=(0× percentage not stained) + (1× percentage weakly stained) + (2× percentage moderately stained) + (3× percentage strongly stained) (34). The mid-point of WH histoscore was used as the cut-off criterion similary as previosly (35,36). MMP9 expression was stratified as low vs. high, according to the cut-off value of WH histoscore (150).

Detecting CTCs in peripheral blood

CTCs were identified via reverse transcription-quantitative (RT-q)PCR analysis. Enrichment of CTC from peripheral blood by depleting CD45+ cells was performed using the Rossette Sep™ kit (15162; Stemcell Technologies, Inc.), as previously described (37,38). Briefly, RNA isolated from CD45-depleted peripheral blood samples were transcribed into cDNA, which was subjected to RT-qPCR analysis to assess the expression levels of epithelial-to-mesenchymal transition (EMT-TF) genes, including TWIST1, SNAIL1, SLUG and ZEB1. Compared with healthy donors, patient samples with higher EMT-TF gene transcript levels were classified as CTC EMT positive, based on the preclinical study and human sample testing. The highest expression values in healthy donors were used as a cut-off value to determine CTC positivity (39).

Statistical analysis

Patient characteristics were summarized using the median (range) values for continuous variables and frequency (percentage) for categorical variables. The distribution of MMP9 histoscore was significantly different from the normal distribution (Shapiro-Wilk test), thus non-parametric tests were used for analyses. Mann-Whitney U test was used to compare the differences in distributions of MMP9 expression between two groups of patients with PBC, whereby MMP9 expression was categorized as absent or present. Fisher's exact test or the χ2 test were used where appropriate.

The median follow-up period was estimated as a median observation time among all patients and among those still alive at the time of their last follow-up. Disease-free survival (DFS) was calculated from the date of CTCs measurement to the date of disease recurrence (locoregional or distant), secondary cancer, death or last follow-up. DFS was estimated using the Kaplan-Meier product limit method and log-rank test. Two-sided P<0.05 was considered to indicate a statistically significant difference. Statistical analyses were performed using NCSS 11 statistical software (2016; NCSS, LLC.; ncss.com/software/ncss).

Results

Patient characteristics

The present study enrolled 318 patients with PBC. The median age of the assessed cohort was 60 years (age range, 25–83 years). The majority of patients had node negative (60.1%) and hormone positive (83.6%) tumors; 48/318 patients (15.1%) had a HER-2/neu amplified status. Patient characteristics are summarized in Table I.

Table I.

Patient characteristics.

Table I.

Patient characteristics.

Characteristicn (%)
All patients318 (100.0)
Histology
  Invasive ductal carcinoma272 (85.5)
  Invasive lobular carcinoma32 (10.1)
  Other histological subtypes14 (4.4)
Grade
  Low and intermediate200 (62.9)
  High grade110 (34.6)
  Unknown8 (2.5)
T stage
  T1218 (68.6)
  T2 and more100 (31.4)
N stage
  N0191 (60.1)
  N1mi10 (3.1)
  N168 (21.4)
  N227 (8.5)
  N319 (6.0)
  Unknown3 (0.9)
Hormone receptor status (cut-off 1%)
  Negative for both52 (16.4)
  Positive for either266 (83.6)
HER2 status
  Negative270 (84.9)
  Positive48 (15.1)
Ki67 status
  <20%189 (59.4)
  ≥ 20%128 (40.3)
  Unknown1 (0.3)
Molecular subtype
  Luminal A166 (52.2)
  Luminal B99 (31.1)
  HER2+13 (4.1)
  Triple negative39 (12.3)
  Unknown1 (0.3)
P53 status
  Negative193 (60.7)
  Positive124 (39.0)
  Unknown1 (0.3)
BCL-2 status
  Negative92 (28.9)
  Positive225 (70.8)
  Unknown1 (0.3)
CTC EP
  Negative235 (73.9)
  Positive27 (8.5)
CTC EMT
  Negative235 (73.9)
  Positive56 (17.6)
CTC any
  Negative235 (73.9)
  Positive83 (26.1)

[i] CTC, circulating tumor cells; EMT, epithelial-to-mesenchymal transition; EP, epithelial-positive.

CTCs detection

To establish overexpression of the EMT-inducing TF gene transcripts in patients with PBC, the expression levels were compared between patient samples and healthy donors, as previously described (39). Among the patient samples, CTCs were detected in 83 patients (26.1%). CTCs with only epithelial markers were detected in the peripheral blood of 34 patients (10.7%), while CTCs with an EMT phenotype were present in 56 patients (17.6%).

Association between MMP9 expression, and patients/tumor characteristic and CTCs

MMP9 protein expression in tumor cells was assessed in all patients (n=318) (Fig. 1). However, pathologists were unable to detect stromal cells in 9/318 tumor tissues due to the small sample size, which only constituted tumor cells. Thus, MMP9 expression in stroma was only assessed in 309 patients. MMP9 expression intensity at least 1+ and higher was detected in 255 samples (80.2%) in breast tumor cells and in 307 samples (99.4%) of tumor associated stroma (P<0.00001). The mean WH ± standard error of the mean (SEM) for MMP9 expression was significantly higher in breast tumor cells compared with tumor associated stroma (132.0±5.2 vs. 50.8±3.7; P<0.00001). The association between MMP9 expression in tumor cells and clinicopathological characteristics, as well as its association with CTCs are presented in Table II. The results demonstrated that elevated MMP9 expression was significantly associated with EP/PR positive breast cancer cells (mean WH ± SEM=137.6±5.6 vs. 103.4±12.8, P=0.011) and low proliferating tumors (Ki67 <20%) (mean WH ± SEM=141.1±6.7 vs. 117.9±8.1, P=0.018), while elevated MMP9 expression in tumor associated stroma was associated with hormone receptor (EP/PR) status (mean WH ± SEM=54.6±4.0 vs. 30.7±9.1, P=0.021) (Table III). In our analysis, there was found any association between MMP9 expression in breast cancer cells, or in tumor associated stroma and CTCs.

Table II.

Association between MMP9 expression in tumour cells, patients, tumour characteristics and circulating tumor cells.

Table II.

Association between MMP9 expression in tumour cells, patients, tumour characteristics and circulating tumor cells.

MMP9 expression in tumor cells

CharacteristicNMeanSEMMedianP-value
MMP9 expression
weighted histoscore318132.05.2150NA
Histology 0.081
  Invasive ductal carcinoma272136.05.6150
  Other46108.613.6100
Grade 0.242
  Low and intermediate200135.16.5150
  High grade110130.18.8110
  Unknown880.032.70
T-stage 0.163
  T1218136.46.3150
  >T1100122.69.3100
N stage 0.468
  N0201135.46.5150
  N+114127.18.7150
  Unknown390.053.6100
Hormone receptor status (cut-off 1%) 0.011
  Negative for both52103.412.8100
  Positive for either266137.65.6150
HER2 status 0.792
  Negative270131.45.6150
  Positive48135.713.4150
Ki67 status (cut-off 20%) 0.018
  <20%189141.06.7170
  ≥20%128117.98.1110
  Unknown1250.092.0250
Molecular subtype 0.0711
  Luminal A166138.67.2160
  Luminal B99134.89.3150
  HER2+13106.225.6100
  Triple negative39102.414.8100
  Unknown1250.092.3250
P53 status 0.632
  Negative193131.16.7150
  Positive124134.18.3150
  Unknown150.092.950
BCL-2 0.229
  Negative92126.09.7110
  Positive225134.06.2150
  Unknown1250.092.7250
CTC EP 0.851
  Negative235134.16.0150
  Positive27138.117.5150
CTC EMT 0.300
  Negative235134.16.1150
  Positive56120.412.4135
CTC any 0.472
  Negative235134.16.1150
  Positive83126.110.2150

[i] CTC, circulating tumor cells; EMT, epithelial-to-mesenchymal transition; NA, not applicable; EP, epithelial-positive.

Table III.

Association between MMP9 expression in stromal cells, patients, tumour characteristics and circulating tumor cells.

Table III.

Association between MMP9 expression in stromal cells, patients, tumour characteristics and circulating tumor cells.

MMP9 expression in stromal cells

CharacteristicNMeanSEMMedianP-value
MMP9 expression
weighted histoscore30950.83.725NA
Histology 0.434
  Invasive ductal carcinoma26651.94.030
  Other4344.29.820
Grade 0.489
  Low and intermediate19450.94.630
  High grade10851.76.220
  Unknown734.324.40
T-stage 0.469
  T121352.64.425
  >T19646.86.625
N stage 0.536
  N019754.94.630
  N+11043.56.120
  Unknown250.045.650
Hormone receptor status (cut-off 1%) 0.021
  Negative for both4930.79.15
  Positive for either26054.64.030
HER2 status 0.872
  Negative26350.74.020
  Positive4651.69.530
Ki67 status (cut-off 20%) 0.137
  <20%18356.34.830
  ≥20%12642.95.720
Molecular subtype 0.094
  Luminal A16357.068.730
  Luminal B9750.562.030
  HER2+121524.30
  Triple negative3735.855.75
P53 status 0.537
  Negative18848.14.730
  Positive12055.55.920
  Unknown10.064.50
BCL-2 0.995
  Negative8847.96.930
  Positive22152.04.320
CTC EP 0.350
  Negative22954.44.130
  Positive2741.312.820
CTC EMT 0.400
  Negative22954.44.130
  Positive5340.31010
CTC any 0.168
  Negative22954.44.130
  Positive8040.67.220

[i] CTC, circulating tumor cells; EP, epithelial; EMT, epithelial-to-mesenchymal transition; NA, not applicable.

Prognostic value of MMP9 in PBC

The median follow-up time was 54.9 months (range, 0.2–76.6 months). In the assessed cohort, 61 patients (19.2%) experienced a disease progression during follow-up. Among the subgroup of patients where MMP9 expression in tumor associated stroma was assessed (n=309), the median follow-up time was 55.3 months (range, 0.2–76.6), and 59 patients (19.1%) experienced a DFS event. Due to insufficiency of overall survival data, only DFS data are presented in the present study.

Univariate analysis was performed to determine the prognostic value of MMP9 in PBC cells [hazard ratio (HR)=0.96; 95% confidence interval (CI), 0.58–1.59; P=0.864; Fig. 2A], as well as in tumor associated stroma (HR=1.29; 95% CI, 0.60–2.78; P=0.547; Fig. 2B). Exploratory subgroup analysis was performed to determine a potential subgroup-related prognostic value of MMP9 (Tables IV and V). In addition, also the univariate analysis in group of patients with concomitant high MMP9 expression in tumor and stromal cells was carried out. However, no prognostic value was found using this analysis (HR=1.27, 95% CI 0.59–2.75, P=0.573) (Fig. 2C). The results demonstrated that low MMP9 expression in tumor cells was associated with better DFS in hormone receptor (ER/PR) negative and triple negative patients with PBC (HR=0.33; 95% CI, 0.12–0.93; P=0.025; Fig. 3A) and (HR=0.17; 95% CI, 0.05–0.57; P=0.003; Fig. 3B), respectively. Notably, the prognostic value of MMP9 in tumor cells was also observed in the CTC_EMT-positive subgroup of patients (HR=0.40; 95% CI, 0.16–0.95; P=0.047; Fig. 3C). Among the subgroup of patients where MMP9 expression in tumor associated stroma was assessed, the prognostic value of MMP9 was observed in the hormone receptor (ER/PR) negative subgroup of patients (HR=0.14; 95% CI, 0.00–4.81; P=0.002; Fig. 4A), triple negative (HR=0.12; 95% CI, 0.00–4.89; P=0.001; Fig. 4B). In addition, among the subgroup of CTC_EMT positive patients was progression of the disease documented in 1 of 2 patients with high MMP9 expression in stromal cells compared to 22 of 51 patients with low MMP9 expression within 4-years follow up. In subgroup of the CTC_EP positive patients 2 of 4 patient with high MMP9 expression in stromal cells experienced progression of disease compared to 6 of 22 patient with low MMP9 expression after 4-years follow up.

Table IV.

Univariate analysis for disease-free survival according to MMP9 expression in tumor cells.

Table IV.

Univariate analysis for disease-free survival according to MMP9 expression in tumor cells.

CharacteristicNHR95% Low CI95% High CIP-value
Overall 0.864
  Low MMP9 expression1560.960.581.59
  High MMP9 expression162
Invasive ductal carcinoma 0.550
  Low MMP9 expression1260.840.481.47
  High MMP9 expression146
Other histology 0.335
  Low MMP9 expression302.120.558.17
  High MMP9 expression16
Intermediate/low gradea 0.901
  Low MMP9 expression930.950.442.07
  High MMP9 expression107
High grade8 pts NA 0.518
  Low MMP9 expression580.800.401.58
  High MMP9 expression52
T1 stage 0.936
  Low MMP9 expression990.970.481.98
  High MMP9 expression119
T2 stage and higher 0.484
  Low MMP9 expression570.770.371.60
  High MMP9 expression43
N0 stageb 0.844
  Low MMP9 expression990.920.412.09
  High MMP9 expression102
N+ stageb 0.909
  Low MMP9 expression551.040.542.00
  High MMP9 expression59
ER/PR positive for either 0.539
  Low MMP9 expression1221.200.662.18
  High MMP9 expression144
ER/PR negative for both 0.025
  Low MMP9 expression340.330.120.93
  High MMP9 expression18
HER positive 0.712
  Low MMP9 expression221.220.423.49
  High MMP9 expression26
HER negative 0.741
  Low MMP9 expression1340.910.511.62
  High MMP9 expression136
Ki67 low (<20%) 0.523
  Low MMP9 expression841.300.572.99
  High MMP9 expression105
Ki67 high (≥20%) 0.149
  Low MMP9 expression720.620.331.19
  High MMP9 expression57
Triple negativec 0.003
  Low MMP9 expression260.170.050.57
  High MMP9 expression13
P53 negativec 0.735
  Low MMP9 expression960.900.491.65
  High MMP9 expression97
P53 positivec 0.829
  Low MMP9 expression591.110.432.82
  High MMP9 expression65
BCL2 negativec 0.124
  Low MMP9 expression510.530.241.18
  High MMP9 expression41
BCL2 positivec 0.445
  Low MMP9 expression1051.290.672.49
  High MMP9 expression120
CTC EP negative 0.387
  Low MMP9 expression1151.330.692.57
  High MMP9 expression120
CTC EP positive 0.675
  Low MMP9 expression121.520.2011.24
  High MMP9 expression15
CTC EMT negative 0.387
  Low MMP9 expression1151.330.692.47
  High MMP9 expression120
CTC EMT positive 0.047
  Low MMP9 expression290.400.160.95
  High MMP9 expression27
CTC any negative 0.387
  Low MMP9 expression1151.330.692.57
  High MMP9 expression120
CTC any positive 0.113
  Low MMP9 expression410.510.231.14
  High MMP9 expression42

a Data not available in 8 patients

b data not available in 3 patients

c data not available in one patient; ER, estrogen receptor; PR, progesterone receptor; CTC, circulating tumor cells; EMT, epithelial-to-mesenchymal transition; EP, epithelial; HZ, hazard ratio; CI, confidence interval; NA, not applicable.

Table V.

Univariate analysis for disease-free survival according to MMP9 expression in stromal cells.

Table V.

Univariate analysis for disease-free survival according to MMP9 expression in stromal cells.

CharacteristicNHR95% Low CI95% High CIP-value
Overall 0.547
  Low MMP9 expression2761.290.602.78
  High MMP9 expression33
Invasive ductal carcinoma 0.458
  Low MMP9 expression2371.410.633.18
  High MMP9 expression29
Other histology 0.825
  Low MMP9 expression390.790.087.84
  High MMP9 expression4
Intermediate/low gradea 0.970
  Low MMP9 expression1741.020.313.38
  High MMP9 expression20
High gradea 0.458
  Low MMP9 expression961.560.574.24
  High MMP9 expression12
T1 stage 0.974
  Low MMP9 expression1891.020.362.90
  High MMP9 expression24
T2 stage and higher 0.457
  Low MMP9 expression871.710.545.43
  High MMP9 expression9
N0 stageb 0.460
  Low MMP9 expression1741.710.535.55
  High MMP9 expression23
N+ stageb 0.871
  Low MMP9 expression1000.920.312.70
  High MMP9 expression10
ER/PR positive for either 0.323
  Low MMP9 expression2291.670.713.88
  High MMP9 expression31
ER/PR negative for both 0.002
  Low MMP9 expression470.140.004.81
  High MMP9 expression2
HER positive 0.242
  Low MMP9 expression393.120.8311.74
  High MMP9 expression7
HER negative 0.900
  Low MMP9 expression2371.060.432.64
  High MMP9 expression26
Ki67 low (<20%) 0.486
  Low MMP9 expression1591.670.505.53
  High MMP9 expression24
Ki67 high (≥20%) 0.679
  Low MMP9 expression1170.800.262.50
  High MMP9 expression9
Triple negative 0.001
  Low MMP9 expression350.120.004.89
  High MMP9 expression2
P53 negativec 0.901
  Low MMP9 expression1711.070.392.92
  High MMP9 expression17
P53 positivec 0.482
  Low MMP9 expression1041.680.505.69
  High MMP9 expression16
BCL2 negative 0.078
  Low MMP9 expression830.350.052.29
  High MMP9 expression5
BCL2 positive 0.238
  Low MMP9 expression1932.000.814.96
  High MMP9 expression28
CTC EP negative 0.143
  Low MMP9 expression2022.761.087.09
  High MMP9 expression27
CTC EP positive 0.053
  Low MMP9 expression230.180.012.75
  High MMP9 expression4
CTC EMT negative 0.143
  Low MMP9 expression2022.761.087.09
  High MMP9 expression27
CTC EMT positive 0.168
  Low MMP9 expression510.370.043.50
  High MMP9 expression2
CTC any negative 0.143
  Low MMP9 expression2022.761.087.09
  High MMP9 expression27
CTC any positive 0.128
  Low MMP9 expression740.440.101.91
  High MMP9 expression6

a Data not available in 7 patients

b data not available in 2 patients

c data not available in patient; ER, estrogen receptor; PR, progesterone receptor; CTC, circulating tumor cells; EP, epithelial; EMT, epithelial-to-mesenchymal transition; HZ, hazard ratio; CI, confidence interval; NA, not applicable.

Notably, combinatorial survival analysis of CTC_EMT and MMP9 expression in tumor cells demonstrated that CTC_EMT positive patients with high MMP9 expression had a significantly shorter DFS compared with CTC_EMT negative patients (P<0.00001; Fig. 5).

Discussion

MMPs represent a large family of proteolytic enzymes of the extracellular matrix that are involved in extracellular matrix degradation, tumor cell invasion, metastasis and angiogenesis (19,4042). The results of the present study demonstrated that elevated MMP9 expression levels in tumor cells and tumor associated stroma were significantly associated with favorable tumor characteristics. Hormone-positive tumors exhibited significantly higher MMP9 expression in tumor cells, as well as in tumor associated stromal cells. In addition, the results demonstrated an association between increased MMP9 expression and low proliferation index of Ki67. Although the role of MMP9 and its association with breast cancer has been extensively studied, data regarding the prognostic value of MMP9 are inconsistent. On one hand, it has been reported that MMP9 expression is associated with a shorter relapse-free survival time in patients with primary breast tumours (26,29,43,44). The association between upregulated MMP9 expression and an increased risk of overall survival and relapse-free survival in breast cancer has also been confirmed via meta-analyses by Song et al (45) and Ren et al (46). Conversely, some studies have identified MMP-9 as a favourable prognostic marker for breast cancer (9,47).

The results of the present study demonstrated a significant association between high MMP9 expression in tumour cells and poor DFS in hormone receptor negative, triple negative, as well as in the CTC_EMT-positive subgroup of patients with early breast cancer. Analysis of stromal cells exhibited this association in the hormone receptor negative and triple negative subgroups of patients.

These results are in concordance with previous studies, confirming the association between MMP9 expression and a shorter progression time, particularly in patients with basal-like or triple negative breast cancer (48,49). Controversy regarding the association between MMP9 expression and clinical outcomes in different types of malignant tumors, including breast cancer, suggests the presence of active and inactive forms of MMP9. MMPs are secreted in the form of inactive proenzymes, whose activation is mediated via different molecular mechanisms (20,21). Thus, the level of active MMP9 in stromal cells and tumor cells may vary, which will subsequently account for the differences in clinical outcomes (50).

Notably, the results of the present study demonstrated the prognostic value of MMP9 in the CTC_EMT-positive subgroup of patients. Generally, ETM is considered a developmental process, facilitating the resistance to apoptosis and increased invasion, and is closely associated with development of a cancer stem cell phenotype (19,51) This machinery can be directly induced by MMPs in the target epithelial cells. Expression of proteases, including MMPs is upregulated during reorganization of ECM in EMT. In addition, the process of MMP-induced EMT has been best characterized in mammary epithelial cells (52,53). According to the results of the present study, there was no significant association between any subpopulations of CTCs and MMP9 expression. Contrary to MMP1, MMP9 does not actively participate in the release of CTCs into the blood stream of patients with PBC (31). However, these changes may result in the resistance to therapy, and development of a cancer stem cell phenotype closely associated with poor DFS. Given the limited treatment options for these subgroups of patients (triple-negative and CTC_EMT-positive PBC), MMP9 may potentially offer a novel therapeutic target. In addition, the results from the combinatorial survival analysis demonstrated that CTC_EMT positive patients with MMP9 expression in tumor cells had a significantly lower DFS compared with CTC_EMT negative patients, suggesting that EMT acts as a negative prognostic marker only in subgroups of patients with high MMP9 expression, while the subgroup of CTC_EMT positive patients, with low MMP9 expression exhibited no effects.

The spectrum of synthetized MMP inhibitors (MMPIs) assessed in clinical trials have demonstrated poor effectiveness and serious side effects (54,55). The limited clinical effect of MMPIs may be due to their poor selectivity. Previous studies have focused on a broad spectrum of MMPs, most of which exert tumorigenic activity. However, it is necessary to take into consideration that some MMPs are characterized by antitumorigenic effects. Another reason for MMPIs inefficiency can be due to their administration to unselected groups of patients (44,56).

In conclusion, this prospective translational study demonstrated the protective role of MMP9 in patients with breast cancer, whereby its increased expression was associated with favourable tumour characteristics. Thus, as it has been proposed by Pozzi et al (57), inhibition of MMP9 antitumorigenic and antiangiogenic activities may result in a paradoxical increase of tumor angiogenesis and tumor growth. Conversely, the results of the present study demonstrated the association between high MMP9 expression and poor DFS in selected subgroups of patients with PBC, particularly hormone receptor negative and triple negative tumors, as well as in CTC_EMT positive patients. These results suggest that MMP9 exerts different biological roles in HR positive vs. negative tumors, further supporting the concept of different biology of breast cancer subtypes according to their HR status. Thus, assessing MMP9 tumor expression may help identify individuals with increased risk of disease recurrence within the aforementioned subgroups of patients with PBC. However, there were certain limitations to the present study, such as the retrospective design of the study and semi-quantitative IHC analysis used for investigating of MMP9 expression. In addition, the study population represent a homogenous cohort of patients, treatment-naïve, without metastatic disease, in order to avoid the effect of the metastatic site heterogeneity factor on analysed variables.

Further studies are required to develop selective MMPIs against the specific protumorigenic MMPs or protumorigenic activities of selected MMPs. Another strategy may be anticancer therapy with antitumorigenic MMPs or with their antitumorigenic subparts. An example of this phenomenon involves the MMP8 enzyme, whereby high MMP8 expression supresses metastasis, while MMP8 silencing induces tumour progression and metastasis (5860).

Acknowledgements

The authors would like to thank Dr Gabriela Sieberova, Dr Jan Macuch, Dr Michal Majercik, Dr Peter Jani and Professor Pavel Babal (all from Department of Pathology, Faculty of Medicine, Comenius University) for their valuable input into IHC evaluation. The authors would also like to thank Ms. Zlatica Pekova (2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute) for administrative support, and Ms. Emilia Klincova and Mr. Ludovit Gaspar (both, Department of Pathology, Faculty of Medicine, Comenius University) for their excellent technical assistance.

Funding

The present study was funded by the Slovak Research and Development Agency (grant no. APVV-16-0010).

Availability of data and materials

All datasets generated and analyzed during the present study are included in this published article.

Authors' contributions

KK, ZC, JM, MM and GM conceived and designed the present study. KK performed statistical analysis. ZC and IM performed immunohistochemical analysis. GM, TS and DK were involved in CTC detection. MK, JB and DP were involved in patient accrual and performed breast surgery. KK and MM drafted the initial manuscript, and all authors reviewed it critically for important intellectual content. All authors participated in the acquisition, analysis and interpretation of data. All authors have read and approved the final version of the manuscript for publication.

Ethics approval and consent to participate

The present study was reviewed and approved by the Institutional Review Board of the National Cancer Institute of Slovakia, Bratislava, Slovakia (approval no. TRU-SK 002; Chair: Professor Michal Mego). Written informed consent was provided by all patients prior to study commencement.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

MMP9

matrix metalloproteinase 9

CTCs

circulating tumor cells

EMT

epithelial-to-mesenchymal transition

ER

estrogen receptor

HR

hazard ratio

PBC

primary breast cancer

PR

progesterone receptor

RT-PCR

reverse transcription PCR

NA

non-applicable

References

1 

Ghoncheh M, Pournamdar Z and Salehiniya H: Incidence and mortality and epidemiology of breast cancer in the world. Asian Pac J Cancer Prev. 17:43–46. 2016. View Article : Google Scholar

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar

3 

Moreau JE, Anderson K, Mauney JR, Nguyen T, Kaplan DL and Rosenblatt M: Tissue-engineered bone serves as a target for metastasis of human breast cancer in a mouse model. Cancer Res. 67:10304–10308. 2007. View Article : Google Scholar

4 

Fagan-Solis KD, Schneider SS, Pentecost BT, Bentley BA, Otis CN, Gierthy JF and Arcaro KF: The RhoA pathway mediates MMP-2 and MMP-9-independent invasive behavior in a triple-negative breast cancer cell line. J Cell Biochem. 114:1385–1394. 2013. View Article : Google Scholar

5 

Malmgren JA, Mayer M, Atwood MK and Kaplan HG: Differential presentation and survival of de novo and recurrent metastatic breast cancer over time: 1990–2010. Breast Cancer Res Treat. 167:579–590. 2018. View Article : Google Scholar

6 

Fan J, Deng X, Gallagher JW, Huang H, Huang Y, Wen J, Ferrari M, Shen H and Hu Y: Monitoring the progression of metastatic breast cancer on nanoporous silica chips. Philos Trans- Royal Soc, Math Phys Eng Sci. 370:2433–2447. 2012.

7 

Bottino J, Gelaleti GB, Maschio LB, Jardim-Perassi BV and de Campos Zuccari DA: Immunoexpression of ROCK-1 and MMP-9 as prognostic markers in breast cancer. Acta Histochem. 116:1367–1373. 2014. View Article : Google Scholar

8 

Olayide A, Samuel O, Ganiyu R, Moses A, Gafar O, Abiola D, Dapo K and John A: How effective is the treatment of locally advanced and metastatic breast cancer in developing centres?: A retrospective review. Ethiop J Health Sci. 25:337–344. 2015. View Article : Google Scholar

9 

Scorilas A, Karameris A, Arnogiannaki N, Ardavanis A, Bassilopoulos P, Trangas T and Talieri M: Overexpression of matrix-metalloproteinase-9 in human breast cancer: A potential favourable indicator in node-negative patients. Br J Cancer. 84:1488–1496. 2001. View Article : Google Scholar

10 

Fidler IJ: Metastasis: Quantitative analysis of distribution and fate of tumor emboli labeled with 125 I-5-iodo-2-deoxyuridine. J Natl Cancer Inst. 45:773–782. 1970.

11 

Lambert AW, Pattabiraman DR and Weinberg RA: Emerging biological principles of metastasis. Cell. 168:670–691. 2017. View Article : Google Scholar

12 

Nguyen DX, Bos PD and Massagué J: Metastasis: From dissemination to organ-specific colonization. Nat Rev Cancer. 9:274–284. 2009. View Article : Google Scholar

13 

Hsiao KC, Shih NY, Fang HL, Huang TS, Kuo CC, Chu PY, Hung YM, Chou SW, Yang YY, Chang GC, et al: Surface α-enolase promotes extracellular matrix degradation and tumor metastasis and represents a new therapeutic target. PLoS One. 8:e693542013. View Article : Google Scholar

14 

Wang X, Lu H, Urvalek AM, Li T, Yu L, Lamar J, DiPersio CM, Feustel PJ and Zhao J: KLF8 promotes human breast cancer cell invasion and metastasis by transcriptional activation of MMP9. Oncogene. 30:1901–1911. 2011. View Article : Google Scholar

15 

Ortíz-López L, Morales-Mulia S, Ramírez-Rodríguez G and Benítez-King G: ROCK-regulated cytoskeletal dynamics participate in the inhibitory effect of melatonin on cancer cell migration. J Pineal Res. 46:15–21. 2009. View Article : Google Scholar

16 

Paolillo M and Schinelli S: Extracellular matrix alterations in metastatic processes. Int J Mol Sci. 20:49472019. View Article : Google Scholar

17 

Castro-Castro A, Marchesin V, Monteiro P, Lodillinsky C, Rossé C and Chavrier P: Cellular and molecular mechanisms of MT1-MMP-dependent cancer cell invasion. Annu Rev Cell Dev Biol. 32:555–576. 2016. View Article : Google Scholar

18 

Shay G, Lynch CC and Fingleton B: Moving targets: Emerging roles for MMPs in cancer progression and metastasis. Matrix Biol. 44-46:200–206. 2015. View Article : Google Scholar

19 

Gonzalez-Avila G, Sommer B, Mendoza-Posada DA, Ramos C, Garcia-Hernandez AA and Falfan-Valencia R: Matrix metalloproteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit Rev Oncol Hematol. 137:57–83. 2019. View Article : Google Scholar

20 

Yousef EM, Tahir MR, St-Pierre Y and Gaboury LA: MMP-9 expression varies according to molecular subtypes of breast cancer. BMC Cancer. 14:6092014. View Article : Google Scholar

21 

Stuelten CH, DaCosta Byfield S, Arany PR, Karpova TS, Stetler-Stevenson WG and Roberts AB: Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-alpha and TGF-beta. J Cell Sci. 118:2143–2153. 2005. View Article : Google Scholar

22 

Sand JM, Larsen L, Hogaboam C, Martinez F, Han M, Larsen MR, Nawrocki A, Zheng Q, Karsdal MA and Leeming DJ: MMP mediated degradation of type IV collagen alpha 1 and alpha 3 chains reflects basement membrane remodeling in experimental and clinical fibrosis - validation of two novel biomarker assays. PLoS One. 8:e849342013. View Article : Google Scholar

23 

Kessenbrock K, Plaks V and Werb Z: Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell. 141:52–67. 2010. View Article : Google Scholar

24 

Owyong M, Chou J, van den Bijgaart RJ, Kong N, Efe G, Maynard C, Talmi-Frank D, Solomonov I, Koopman C, Hadler-Olsen E, et al: MMP9 modulates the metastatic cascade and immune landscape for breast cancer anti-metastatic therapy. Life Sci Alliance. 2:e2018002262019. View Article : Google Scholar

25 

Li HC, Cao DC, Liu Y, Hou YF, Wu J, Lu JS, Di GH, Liu G, Li FM, Ou ZL, et al: Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 88:75–85. 2004. View Article : Google Scholar

26 

Vizoso FJ, González LO, Corte MD, Rodríguez JC, Vázquez J, Lamelas ML, Junquera S, Merino AM and García-Muñiz JL: Study of matrix metalloproteinases and their inhibitors in breast cancer. Br J Cancer. 96:903–911. 2007. View Article : Google Scholar

27 

Darlix A, Lamy PJ, Lopez-Crapez E, Braccini AL, Firmin N, Romieu G, Thézenas S and Jacot W: Serum NSE, MMP-9 and HER2 extracellular domain are associated with brain metastases in metastatic breast cancer patients: Predictive biomarkers for brain metastases? Int J Cancer. 139:2299–2311. 2016. View Article : Google Scholar

28 

Ranogajec I, Jakić-Razumović J, Puzović V and Gabrilovac J: Prognostic value of matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9) and aminopeptidase N/CD13 in breast cancer patients. Med Oncol. 29:561–569. 2012. View Article : Google Scholar

29 

Li H, Qiu Z, Li F and Wang C: The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 14:5865–5870. 2017.

30 

Dhar M, Lam JN, Walser T, Dubinett SM, Rettig MB and Di Carlo D: Functional profiling of circulating tumor cells with an integrated vortex capture and single-cell protease activity assay. Proc Natl Acad Sci USA. 115:9986–9991. 2018. View Article : Google Scholar

31 

Cierna Z, Mego M, Janega P, Karaba M, Minarik G, Benca J, Sedlácková T, Cingelova S, Gronesova P, Manasova D, et al: Matrix metalloproteinase 1 and circulating tumor cells in early breast cancer. BMC Cancer. 14:4722014. View Article : Google Scholar

32 

Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, Zackrisson S and Senkus E; ESMO Guidelines Committee, : Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 30:16742019. View Article : Google Scholar

33 

Allison KH, Brogi E and Ellis IO: WHO Classification of Tumours Editorial Board. Breast Tumours. 5th edition. IARC Press; Lyon: 2019

34 

van Nes JG, de Kruijf EM, Putter H, Faratian D, Munro A, Campbell F, Smit VT, Liefers GJ, Kuppen PJ, van de Velde CJ, et al: Co-expression of SNAIL and TWIST determines prognosis in estrogen receptor-positive early breast cancer patients. Breast Cancer Res Treat. 133:49–59. 2012. View Article : Google Scholar

35 

Chovanec M, Cierna Z, Miskovska V, Machalekova K, Svetlovska D, Kalavska K, Rejlekova K, Spanik S, Kajo K, Babal P, et al: Prognostic role of programmed-death ligand 1 (PD-L1) expressing tumor infiltrating lymphocytes in testicular germ cell tumors. Oncotarget. 8:21794–21805. 2017. View Article : Google Scholar

36 

Chovanec M, Cierna Z, Miskovska V, Machalekova K, Kalavska K, Rejlekova K, Svetlovska D, Macak D, Spanik S, Kajo K, et al: β-catenin is a marker of poor clinical characteristics and suppressed immune infiltration in testicular germ cell tumors. BMC Cancer. 18:10622018. View Article : Google Scholar

37 

Mego M, Karaba M, Minarik G, Benca J, Sedlácková T, Tothova L, Vlkova B, Cierna Z, Janega P, Luha J, et al: Relationship between circulating tumor cells, blood coagulation, and urokinase-plasminogen-activator system in early breast cancer patients. Breast J. 21:155–160. 2015. View Article : Google Scholar

38 

Mego M, Karaba M, Minarik G, Benca J, Silvia J, Sedlackova T, Manasova D, Kalavska K, Pindak D, Cristofanilli M, et al: Circulating tumor cells with epithelial-to-mesenchymal transition phenotypes associated with inferior outcomes in primary breast cancer. Anticancer Res. 39:1829–1837. 2019. View Article : Google Scholar

39 

Mego M, Mani SA, Lee BN, Li C, Evans KW, Cohen EN, Gao H, Jackson SA, Giordano A, Hortobagyi GN, et al: Expression of epithelial-mesenchymal transition-inducing transcription factors in primary breast cancer: The effect of neoadjuvant therapy. Int J Cancer. 130:808–816. 2012. View Article : Google Scholar

40 

Yadav L, Puri N, Rastogi V, Satpute P, Ahmad R and Kaur G: Matrix metalloproteinases and cancer - roles in threat and therapy. Asian Pac J Cancer Prev. 15:1085–1091. 2014. View Article : Google Scholar

41 

Westermarck J and Kähäri VM: Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J. 13:781–792. 1999. View Article : Google Scholar

42 

Alaseem A, Alhazzani K, Dondapati P, Alobid S, Bishayee A and Rathinavelu A: Matrix metalloproteinases: A challenging paradigm of cancer management. Semin Cancer Biol. 56:100–115. 2019. View Article : Google Scholar

43 

González LO, Pidal I, Junquera S, Corte MD, Vázquez J, Rodríguez JC, Lamelas ML, Merino AM, García-Muñiz JL and Vizoso FJ: Overexpression of matrix metalloproteinases and their inhibitors in mononuclear inflammatory cells in breast cancer correlates with metastasis-relapse. Br J Cancer. 97:957–963. 2007. View Article : Google Scholar

44 

Radisky ES, Raeeszadeh-Sarmazdeh M and Radisky DC: Therapeutic Potential of Matrix Metalloproteinase Inhibition in Breast Cancer. J Cell Biochem. 118:3531–3548. 2017. View Article : Google Scholar

45 

Song J, Su H, Zhou YY and Guo LL: Prognostic value of matrix metalloproteinase 9 expression in breast cancer patients: A meta-analysis. Asian Pac J Cancer Prev. 14:1615–1621. 2013. View Article : Google Scholar

46 

Ren F, Tang R, Zhang X, Madushi WM, Luo D, Dang Y, Li Z, Wei K and Chen G: Overexpression of MMP family members functions as prognostic biomarker for breast cancer patients: A systematic review and meta-analysis. PLoS One. 10:e01355442015. View Article : Google Scholar

47 

Pellikainen JM, Ropponen KM, Kataja VV, Kellokoski JK, Eskelinen MJ and Kosma VM: Expression of matrix metalloproteinase (MMP)-2 and MMP-9 in breast cancer with a special reference to activator protein-2, HER2, and prognosis. Clin Cancer Res. 10:7621–7628. 2004. View Article : Google Scholar

48 

González LO, Corte MD, Junquera S, González-Fernández R, del Casar JM, García C, Andicoechea A, Vázquez J, Pérez-Fernández R and Vizoso FJ: Expression and prognostic significance of metalloproteases and their inhibitors in luminal A and basal-like phenotypes of breast carcinoma. Hum Pathol. 40:1224–1233. 2009. View Article : Google Scholar

49 

Zhao S, Ma W, Zhang M, Tang D, Shi Q, Xu S, Zhang X, Liu Y, Song Y, Liu L, et al: High expression of CD147 and MMP-9 is correlated with poor prognosis of triple-negative breast cancer (TNBC) patients. Med Oncol. 30:3352013. View Article : Google Scholar

50 

Yang J, Min KW, Kim DH, Son BK, Moon KM, Wi YC, Bang SS, Oh YH, Do SI, Chae SW, et al: High TNFRSF12A level associated with MMP-9 overexpression is linked to poor prognosis in breast cancer: Gene set enrichment analysis and validation in large-scale cohorts. PLoS One. 13:e02021132018. View Article : Google Scholar

51 

Radisky ES and Radisky DC: Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia. 15:201–212. 2010. View Article : Google Scholar

52 

Foroni C, Broggini M, Generali D and Damia G: Epithelial-mesenchymal transition and breast cancer: Role, molecular mechanisms and clinical impact. Cancer Treat Rev. 38:689–697. 2012. View Article : Google Scholar

53 

Li W, Li S, Deng L, Yang S, Li M, Long S, Chen S, Lin F and Xiao L: Decreased MT1-MMP in gastric cancer suppressed cell migration and invasion via regulating MMPs and EMT. Tumour Biol. 36:6883–6889. 2015. View Article : Google Scholar

54 

Zhong Y, Lu YT, Sun Y, Shi ZH, Li NG, Tang YP and Duan JA: Recent opportunities in matrix metalloproteinase inhibitor drug design for cancer. Expert Opin Drug Discov. 13:75–87. 2018. View Article : Google Scholar

55 

Arkadash V, Yosef G, Shirian J, Cohen I, Horev Y, Grossman M, Sagi I, Radisky ES, Shifman JM and Papo N: Development of high affinity and high specificity inhibitors of matrix metalloproteinase 14 through computational design and directed evolution. J Biol Chem. 292:3481–3495. 2017. View Article : Google Scholar

56 

Winer A, Adams S and Mignatti P: Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures intofuture successes. Mol Cancer Ther. 17:1147–1155. 2018. View Article : Google Scholar

57 

Pozzi A, LeVine WF and Gardner HA: Low plasma levels of matrix metalloproteinase 9 permit increased tumor angiogenesis. Oncogene. 21:272–281. 2002. View Article : Google Scholar

58 

Montel V, Kleeman J, Agarwal D, Spinella D, Kawai K and Tarin D: Altered metastatic behavior of human breast cancer cells after experimental manipulation of matrix metalloproteinase 8 gene expression. Cancer Res. 64:1687–1694. 2004. View Article : Google Scholar

59 

Decock J, Hendrickx W, Thirkettle S, Gutiérrez-Fernández A, Robinson SD and Edwards DR: Pleiotropic functions of the tumor- and metastasis-suppressing matrix metalloproteinase-8 in mammary cancer in MMTV-PyMT transgenic mice. Breast Cancer Res. 17:382015. View Article : Google Scholar

60 

Juurikka K, Butler GS, Salo T, Nyberg P and Åström P: The role of MMP8 in cancer: A systematic review. Int J Mol Sci. 20:45062019. View Article : Google Scholar

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kalavska K, Cierna Z, Karaba M, Minarik G, Benca J, Sedlackova T, Kolekova D, Mrvova I, Pindak D, Mardiak J, Mardiak J, et al: Prognostic role of matrix metalloproteinase 9 in early breast cancer. Oncol Lett 21: 78, 2021
APA
Kalavska, K., Cierna, Z., Karaba, M., Minarik, G., Benca, J., Sedlackova, T. ... Mego, M. (2021). Prognostic role of matrix metalloproteinase 9 in early breast cancer. Oncology Letters, 21, 78. https://doi.org/10.3892/ol.2020.12339
MLA
Kalavska, K., Cierna, Z., Karaba, M., Minarik, G., Benca, J., Sedlackova, T., Kolekova, D., Mrvova, I., Pindak, D., Mardiak, J., Mego, M."Prognostic role of matrix metalloproteinase 9 in early breast cancer". Oncology Letters 21.2 (2021): 78.
Chicago
Kalavska, K., Cierna, Z., Karaba, M., Minarik, G., Benca, J., Sedlackova, T., Kolekova, D., Mrvova, I., Pindak, D., Mardiak, J., Mego, M."Prognostic role of matrix metalloproteinase 9 in early breast cancer". Oncology Letters 21, no. 2 (2021): 78. https://doi.org/10.3892/ol.2020.12339