Open Access

A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment

  • Authors:
    • Mingyue Li
    • Shugang Xing
    • Haiying Zhang
    • Siqi Shang
    • Xiangxiang Li
    • Bo Ren
    • Gaiyun Li
    • Xiaona Chang
    • Yilei Li
    • Wei Li
  • View Affiliations

  • Published online on: January 5, 2016     https://doi.org/10.3892/or.2016.4547
  • Pages: 1329-1339
  • Copyright: © Li 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

Anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) treatment is effective for the treatment of primary tumors, but not sufficient for the treatment of metastatic tumors, likely owing to the effects of the tumor microenvironment. In this study, we aimed to determine the therapeutic effects of combined treatment with a matrix metalloproteinase (MMP) inhibitor (MMPI) and anti-CTLA-4 antibody in a breast cancer model in mice. Interestingly, combined treatment with MMPI and anti-CTLA-4 antibody delayed tumor growth and reduced lung and liver metastases compared with anti-CTLA-4 alone or vehicle treatment. The functions of the liver and kidney in mice in the different groups did not differ significantly compared with that in normal mice. The CD8+/CD4+ ratio in T cells in the spleen and tumor were increased after monotherapy or combined anti-CTLA-4 antibody plus MMPI therapy compared with that in vehicle-treated mice. Anti-CTLA-4 antibody plus MMPI therapy reduced the percentage of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) and decreased the Treg/Th17 cell ratio in the spleen compared with those in the vehicle-treated group. Additionally, anti-CTLA-4 antibody plus MMPI therapy reduced the percentages of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and Th17 cells in tumors compared with that in the vehicle-treated group. Moreover, combined treatment with MMPI and anti-CTLA-4 antibody reduced the microvessel density (MVD) in tumors compared with that in vehicle or MMPI-treated mice. There was a negative correlation between MVD and the CD8+ T cell percentage, CD4+ T cell percentage, and CD8+/CD4+ T cell ratio, but a positive correlation with Tregs, Th17 cells, Treg/Th17 cell ratio, and MDSCs. Thus, these data demonstrated that addition of MMPI enhanced the effects of anti-CTLA-4 antibody treatment in a mouse model of breast cancer by delaying tumor growth and reducing metastases.

Introduction

Cytotoxic T lymphocyte antigen-4 (CTLA-4) is an inhibitory molecule found on T cells. Immunotherapy by blocking CTLA-4 can enhance T-cell activation and proliferation and improve antitumor immune responses (1). However, anti-CTLA-4 antibody therapy is not sufficient for the treatment of metastatic malignant tumors (2), likely owing to the effects of the tumor microenvironment (TME) (3). The TME has a more stable genetic background and has been shown to participate in regulating tumor immune escape and mediating the sensitivity of tumor cells to anticancer drugs (47). Matrix metalloproteinases (MMPs) are the primary factor regulating the TME through degradation of the extracellular matrix and promotion of tumor angiogenesis (8). Moreover, MMPs play a key role in promoting the occurrence and development of cancer (9). However, the specific effects of combined inhibition of CTLA-4 and MMPs in breast cancer are unknown.

Therefore, in this study, we constructed a breast cancer model in mice using a highly metastatic breast cancer cell line. We used this model to evaluate the therapeutic effects of anti-CTLA-4 antibody therapy alone or combination with an MMP inhibitor (MMPI) and to explore the role of the TME in tumor treatment.

Materials and methods

Murine mammary carcinoma cells and the animal model

The 4T1 murine mammary carcinoma cell line harboring the luciferase construct was provided by the Pathology Department of the College of Basic Medical Sciences, Jilin University, China. 4T1 cells were cultured in RPMI-1640 medium (Gibco, USA) supplemented with 10% fetal bovine serum (FBS; Hyclone, USA) and incubated at 37°C in an atmosphere containing 5% CO2. In the logarithmic growth phase, 4T1 cells were collected and diluted to a concentration of 5×106 cells/ml with phosphate-buffered saline (PBS). Next, 0.2 ml of the cell suspension was injected subcutaneously into the right flanks of 6–8-week-old female BALB/c mice, provided by the Laboratory Animal Center of the College of Basic Medical Sciences, Jilin University. All procedures followed the animal care regulations of the University Health Network and were approved by the Research Ethic Board of Jilin University.

Treatment of tumor-bearing mice

Tumor nodules reached 50 mm3 on day 3. Mice were randomly divided into four groups as follows: vehicle, anti-CTLA-4 antibody alone (a-CTLA-4); MMPI alone, and combination therapy (a-CTLA-4 plus MMPI). Mice were treated with 100 µg anti-CTLA-4 antibody (clone, 9H10; BioXCell, West Lebanon, NH, USA) by intraperitoneal (i.p.) injection once every two day and/or with 0.1 MMPI (1 mg/kg) by subcutaneous injection once every two days. The MMPI potassium ferricyanide {K3[Fe(CN)6 (10,11), was kindly provided by Professor Xuexun Fang (Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, Jilin University, China). Mice in the vehicle group were treated with an equal volume of normal saline and PBS. The dosages used in the combined treatment group were equal to those used in the monotherapy groups. The longest (a) and shortest (b) diameters of the tumor were measured with calipers every 3 days, and tumor volume was estimated by the formula V = ab2/2. On days 7, 21 and 35, tumors were measured using an Ultrasound Biomicroscopy InviVue instrument (PanoView β1500; Taiwan).

Biophotonic imaging of animals

Mice were anesthetized by i.p. injection of 4% chloral hydrate and then subjected to i.p. injection with 200 µl D-luciferin (15 mg/ml; GoldBio, St. Louis, MO, USA). After 10 min, the mice were imaged using a small animal in vivo optical imaging system (IVIS Spectrum; Caliper Life Sciences, USA).

Hematoxylin and eosin (H&E) staining

Mice were sacrificed. The lungs and livers were immediately placed in Bouin's fixation and 10% formaldehyde. After 48 h, metastatic lesions on the surface of the lungs (faint yellow in color) were counted without the use of a microscope. The lungs and livers of each mouse were then embedded in paraffin, sectioned, stained with H&E staining, and examined histologically for evidence of metastatic lesions within the lung and liver tissue. For each lung and liver, three consecutive sections, separated by 200 µm, were collected. The sections was randomized and coded, and the total number of metastatic foci was counted.

Functional tests for the liver and kidneys

Peripheral blood was obtained from each mouse and centrifuged at 500 × g for 20 min after standing at room temperature for 20 min. The supernatant was collected as serum. Total protein, serum albumin, serum globulin, albumin/globulin, alanine aminotransferase (ALT), aspartate aminotransferase (AST), ALT/AST, alkaline phosphatase, urea, creatinine, and uric acid in serum were measured using an automatic biochemical analyzer (Beckman Coulter, USA).

Flow cytometry analysis

The spleens of mice were placed in cold RPMI-1640 medium immediately and then cut into small pieces with eye scissors. Single-cell suspensions of spleen tissues were acquired after filtration with 300- and 100-mesh filters. Single-cell suspensions of bone marrow tissues were obtained from the thigh bones of mice after being washed with cold PBS. The red blood cells were then disrupted within the cell suspension. For treatment, 2×106 cells were stimulated with phorbol 12-myristate 13-acetate (PMA; Sigma, St. Louis, MO, USA; 50 ng/ml) plus 2 µg/ml ionomycin (Sigma) and monensin (Sigma; 5 µg/ml) for 5 h. The cells were then collected for Th17 cytokine staining. Subsequently, they were fixed and/or permeabilized using fixation/permeabilization kits (BD, USA) according to the manufacturer's instructions, and incubated with anti-interleukin (IL)-17A-Alexa Fluor (BD), anti-CD4-PE (BD), anti-CD4-APC (BD), anti-CD25-PE-CyTM7 (BD), anti-FoxP3-PE (eBioscience, San Diego, CA, USA), anti-CD3e-FITC (BD), anti-CD4-PE-CyTM7 (BD), anti-CD8-PE (BD), anti-CD11b-APC (BD), and anti-Gr-1-PE (BD) antibodies for 40 min at 4°C. The cells were incubated with rat anti-mouse CD16/CD32 (BD) for 30 min before CD11b and Gr-1 staining at 4°C. Flow cytometry (BD) was performed, and data were analyzed using FlowJo software.

Immunohistochemistry analysis

For immunohistochemical analysis of microvessel density (MVD), 2-µm-thick sections of formalin-fixed, paraffin-embedded mammary tumor tissue sections were deparaffinized in xylene and rehydrated in graded alcohols. For antigen retrieval, slides were immersed and boiled for 5 min in citrate buffer (pH 6.0). Slides were incubated with a peroxidase inhibitor and horse serum blocking solution for 30 min and then subjected to overnight staining with rat anti-mouse CD34 primary monoclonal antibodies (1:100 dilution; ab81289; Abcam, USA) at 4°C. Slides were then washed three times with PBS-T, incubated with horse anti-rat secondary antibodies for 30 min at room temperature, washed again with PBS-T three times, and incubated in DAB peroxide substrate solution. Counterstaining was performed with Harris hematoxylin counterstain. MVD was determined as the mean number of microvessels under five microscopic fields (200×).

Immunofluorescence analysis

Immunofluorescence staining of slides was performed as described for immunohistochemistry until antigen retrieval. Then, slides were treated with or without 1% Triton X-100 for 20 min, followed by blocking with 5% bovine serum albumin (BSA) for 1 h at room temperature. Subsequently, slides were incubated with anti-CD3e-FITC (BD), anti-CD8a-PE (BD), anti-CD4-PE (BD), anti-CD11b-FITC (BD), anti-Gr-1-PE (BD), anti-Foxp3-PE (BD), and anti-IL-17A-Alexa Fluor 647 (BD) for 30 min at room temperature. Slides were then washed three times with PBS-T, incubated with Hoechst-33342 for 5 min at room temperature, washed again with PBS-T three times, and sealed with glycerin.

Statistical analysis

All values were expressed as means ± standard deviations (SDs). All statistical analyses were performed using Student's t-tests, non-parametric tests, or Spearman correlation analyses. Differences or correlations with P-values of ≤0.05 were considered significant.

Results

Addition of the MMPI enhances the inhibitory effects of the anti-CTLA-4 antibody on breast cancer growth

First, we established a breast cancer model by subcutaneous inoculation of 4T1 cells in order to easily monitor tumor growth using ultrasound and calipers. By day 3, tumor nodules reached ~50 mm3; therefore, treatments, including anti-CTLA-4 antibody and MMPI, were applied beginning on day 3. Our results demonstrated that treatment with the MMPI or MMPI plus anti-CTLA-4 antibody inhibited the growth of transplanted tumors compared with vehicle or anti-CTLA-4 antibody alone to varying degrees. Indeed, according to analysis of the total days, combined treatment caused a significant reduction in the growth of breast tumors compared with that in vehicle-treated mice (P<0.05; Fig. 1A). Additionally, ultrasonography on days 7, 21 and 35 revealed that tumors in mice treated with anti-CTLA-4 antibody and MMPI were significantly smaller than those in mice treated with vehicle, MMPI alone, or anti-CTLA-4 antibody alone (Fig. 1B). However, the combination treatment had no effect on the survival of tumor-bearing mice (P>0.05; Fig. 1C).

Treatment with the MMPI enhances the effects of the anti-CTLA-4 antibody on breast cancer metastasis

In order to visualize tumor metastasis in live mice, we constructed a mouse model of breast cancer using 4T1 cells stably expressing luciferase. D-luciferin was applied on day 42 for detection of tumor metastasis using a small animal in vivo optical imaging system. The results showed that the mice in the vehicle group had metastatic lesions, whereas the mice in the other groups had no metastatic lesions (Fig. 2A).

Observation of fixed lungs showed that the number of tumor metastases on the lung surface was significantly reduced after combined treatment (P<0.05; Fig. 2B). However, there was no significant reduction after treatment with anti-CTLA-4 antibody alone or MMPI alone (P>0.05; Fig. 2B). Additionally, analysis of paraffin-embedded lung and liver sections stained with H&E revealed that both tissues had reduced numbers of metastases after the combined treatment (P<0.001; Fig. 2C). However, there was no significant difference in the number of metastases between the anti-CTLA-4 antibody group and vehicle group (Fig. 2C).

Effects of the MMPI and anti-CTLA-4 antibody on the function of the liver and kidney

MMPIs have been reported to have toxic effects. Therefore, in this study, we examined changes in liver and kidney function in mice after treatment with the MMPI. The results showed that the liver and kidney functions of tumor-bearing mice treated with the MMPI did not differ from those of control mice (P>0.05; Fig. 3), indicating that the MMPI used in this study did not damage the liver or kidneys of mice.

Treatment with the MMPI and anti-CTLA-4 antibody improves the immune microenvironment in mice

Our data showed that MMPI could enhance the therapeutic effects of anti-CTLA 4 antibodies in a model of breast cancer in mice. The mechanism through which the anti-CTLA-4 antibody improves the antitumor immune response involves prompt T-cell activation and proliferation. Therefore, in order to determine whether the MMPI affected the immune microenvironment of tumor-bearing mice to enhance the therapeutic effects of the anti-CTLA-4 antibody, we used flow cytometry to measure changes in the percentages of CD4+ T cells, CD8+ T cells, Tregs, Th17 cells, and MDSCs in the spleens of mice and to evaluate changes of MDSCs in the bone marrow. The results showed that there were no significant differences in the number of CD4+ and CD8+ T cells between the groups (Fig. 4). However, the ratio of CD8+ T cells to CD4+ T cells was significantly increased after MMPI treatment (P<0.05; Fig. 4). Moreover, combined treatment caused significant decreases in Tregs and the Treg/Th17 ratio in mouse spleens (P<0.01; Fig. 4), and the percentage of MDSCs in spleens and the bone marrow was significantly reduced after combination treatment (P<0.01; Fig. 4).

Thus, taken together, these data suggested that the anticancer mechanism of the MMPI may be related to the increased proportion of CD8+ T cells, relieving immune suppression and enhancing the antitumor function of the immune system.

Treatment with the MMPI and anti-CTLA-4 antibody improves the TME

The primary target of MMPIs is the TME, which is generally enriched in immunosuppressive cells. Thus, we examined the infiltration of immune cells into tumor tissues using immunofluorescence. The results showed that while there were no obvious increases in the percentage of CD4+ and CD8+ T cells in the TME, the ratio of CD8+ T cells to CD4+ T cells was increased after MMPI treatment and after combined treatment as compared with that in vehicle-treated mice (P<0.05; Fig. 5). Moreover, the percentages of Tregs, Th17 cells, and MDSCs in the tumors were significantly decreased after combined treatment (P<0.05 for Tregs and Th17 and P<0.01 for MDSCs; Fig. 5). However, the Treg/Th17 ratio did not differ significantly compared with that in vehicle-treated mice. Therefore, these data showed that immunosuppression of the TME was improved to a certain extent after combination treatment with the MMPI and anti-CTLA-4 antibody.

Combined treatment with the MMPI and anti-CTLA-4 antibody reduced the MVD within the TME

Tumor vessels are important elements of the TME, and MVD within the tumor is often associated with tumor progression. We selected paraffin-embedded sections of tumors, stained them with anti-CD34 antibodies to mark tumor stromal vascular endothelial cells, and determined the MVD. Immunohistochemical staining showed that the MVD was higher in tumors from vehicle-treated mice than in tumors from mice treated with the MMPI and anti-CTLA-4 antibody (P<0.01; Fig. 6). Thus, the MMPI may reduce immunosuppression and inhibit neovascularization in the TME.

Correlation between immune cell and MVD within the TME

Immune cells are associated with angiogenesis in the tumor stroma (12). Therefore, we next analyzed the relationship between infiltration of immune cells and MVD within the tumor tissue. Spearman correlation analyses showed that CD8+ T cells, CD4+ T cells, and the CD8+/CD4+ T cell ratio were negatively correlated with MVD in tumor tissues (correlation coefficients: −0.800, −0.800, and −0.100, respectively; P>0.05), whereas Tregs, the Treg/Th17 ratio, Th17 cells, and MDSCs were positively correlated with MVD (correlation coefficients: 0.400, 0.400, 0.800 and 0.800); the correlation between MDSCs and MVD was statistically significant (P<0.05; Fig. 7).

Discussion

In this study, we examined the therapeutic effects of combined treatment with an MMPI and anti-CTLA-4 antibody in a breast cancer model in mice. Our data demonstrated that addition of MMPI enhanced the effects of anti-CTLA-4 antibody treatment in a mouse model of breast cancer by delaying tumor growth and reducing metastases. These results may have important implications in the development of novel therapeutic strategies for the treatment of breast cancer.

Murine breast cancer 4T1 cell can spontaneously metastasize to other organs after subcutaneous injection into female BALB/c mice. This process is similar to the occurrence and development of human breast cancer; therefore, transplantation of 4T1 cells into mice is considered an ideal model for studying human breast cancer (13). The application of anti-CTLA-4 antibodies alone is ineffective for the treatment of breast cancer in mice (14). Therefore, successful immunotherapy using anti-CTLA-4 antibodies would require identification of the appropriate combination therapy. Recent studies have shown that the TME mediates immune escape and regulates the sensitivity of tumors to anticancer drugs; therefore, targeting of the TME may represent a novel method for the treatment of malignant tumors.

MMPs play an important role in tumor progression by regulating the TME. MMPs promote angiogenesis, tumor growth, and tumor spread through the degradation of extracellular matrix, thereby changing the adhesion between cells, promoting cell movement, and regulating the immune response of the tumor (15). Based on the overexpression of MMPs in tumor tissues and the overactivation of MMPs observed in tumors (16), application of an MMPI could block the activity of MMPs, improve the TME, and inhibit tumor growth (17). Indeed, many animal experiments and clinical trials have shown that MMPIs have different degrees of inhibitory effects on tumor growth but are often accompanied by obvious side effects (18,19). Therefore, identification of an MMPI with low toxicity and high efficacy is necessary. In the present study, we used the MMPI potassium ferricyanide, a member of a class of inorganic compounds identified through enzyme kinetics experiments in a previous study. This compound exhibits low toxicity, has weak stimulation in the skin and eyes, and produces toxic gases only when exposed the strong acids. Potassium ferricyanide specifically inhibits MMP-14, MMP-2, and MMP-13 through non-competitive inhibition. Our results of combination therapy with the MMPI (potassium ferricyanide) and anti-CTLA-4 antibody showed that anti-CTLA-4 antibody alone exhibited poor efficacy, consistent with a study by Demaria et al (14), combined treatment with the MMPI and anti-CTLA-4 antibody significantly inhibited the growth and metastasis of breast cancer. Therefore, inhibition of MMPs may improve the therapeutic effects of the anti-CTLA-4 antibody.

Tumor cells are often able to evade immune surveillance and induction of immune tolerance and are thought to be associated with various types of immune cells, including MDSCs, Tregs, and Th17 cells (2022). The antitumor immune response is markedly suppressed by immunosuppressive cells within the body; these cells tend to be found in tumors after treatment with an immunological stimulus, facilitating the development of a microenvironment promoting tumor immunosuppression (23). Tregs play a role in immunosuppression mainly through CTLA-4 on the cell surface (24). MDSCs can produce arginine to inhibit T-cell function and can secrete transforming growth factor (TGF)-β and IL-10 to inhibit T-cell activation (25). Our data suggested that the percentage of Tregs and MDSCs was reduced in spleens and tumors after the combination treatment. Tregs are primarily developed from naïve CD4+ T cells, and TGF-β is the key factor involved in Treg development; indeed, TGF-β can induce the expression of Foxp3, a transcription factor that facilitates the transformation of naïve CD4+ T cells into Tregs (26). MMP-14 induces TGF-β1 protein expression (27); therefore, MMPI may regulate the expression of TGF-β, thus reducing the number of Tregs (28). Additionally, the growth of MDSCs, a large group of naïve cells derived from the bone marrow, can be inhibited by MMP-2 through upregulation of a number of immune suppressor genes, including IL-10, IL-14, IL-11, and chemokine ligand CCL-5 (29). Because the MMPI used in our study could inhibit the activity of MMP-2, we expect that the MMPI may have inhibited the growth of MDSCs, thereby reducing MDSC numbers. Our data also suggested that MDSCs in bone marrow was declined after combined treatment. It was reported that TGF-β signaling pathway is an important factor in regulation of bone marrow-derived MDSCs (28,30). Thus, the inhibition of MMPs may have influence on mature MDSCs. Exosomes may be another factor by which tumor exosomes can switch the differentiation pathway of myeloid cells to the MDSC pathway and of which tumor exosomes will reduce after treatment (31).

Blood vessels in tumors are an important part of the TME and can support the growth of tumors, promote the spread of the tumor cells, and regulate the TME. The tumor vasculature is abnormal, exhibiting circuity and expansion in structure, morphological and structural abnormalities in perithelial cells that are loosely connection or even absent, an incomplete basement membrane, and increased MVD (33). The abnormal blood vessel formation can create an abnormal TME, further affecting the proliferation, invasion, survival, and function of immune cells (33,34). Therefore, the TME is generally characterized by a lack of antitumor immune cells and the accumulation of immunosuppressive cells. Additionally, sustained low concentrations of anti-angiogenic agents can reduce the tumor vascular density, reshape the abnormal blood vessels in the TME, balance vascular perfusion, increase the effects of T-cell infiltration, and improve the immunosuppressive effects of the TME (35,36).

Our data showed that the anti-CTLA-4 antibody alone could not effectively reduce the MVD in tumors; however, combined treatment with the MMPI and anti-CTLA-4 antibody significantly reduced the MVD in tumor tissues. MMPs can promote angiogenesis by degrading the vascular basement membrane, thereby regulating angiogenic factors. MMP-14 plays an important role within the TME and affects angiogenesis by regulating the expression of vascular endothelial growth factor (VEGF) and the biological activities of TGF-β (37). Therefore, the MMPI used in this study may inhibit the angiogenesis-promoting effects of MMPs, thereby reducing the MVD in the TME. In addition, immune cells, particularly immunosuppressive cells, regulate angiogenesis in the TME. Our data suggested that Tregs, Th17 cells, and MDSCs were positively correlated with MVD in the TME; these cells may promote tumor angiogenesis (12). Moreover, hypoxia can promote the infiltration of Tregs into the TME by mediating VEGF-A expression, subsequently promoting endothelial cell recruitment and amplification (38). In ovarian cancer, Tregs are an important source of VEGF in the TME, and removal of Tregs can effectively reduce the production of VEGF. Additionally, Tregs are required for angiogenesis in lung tissue (39). In an alternatively pathway, MDSCs stimulate the activation of signal transducer and activator of transcription 3 (STAT3) in tumor cells via secretion of IL-28 and induction of tumor-associated factors. The tumor cells then promote the formation of tubular structures by endothelial cells (40). Importantly, MDSCs are factors involved in mediating the effects of anti-angiogenic therapy in cancer (41). Angiogenesis is also affected by the expression of IL-17A, which is secreted by Th17 cells and is positively associated with the MVD in gastric cancer, liver cancer, and breast cancer (42). IL-17A indirectly participates in angiogenesis by promoting the secretion of VEGF, prostaglandin E2 (PGE2), and chemotactic factors from tumor cells. IL-17A can also directly affect endothelial cells and endothelial progenitor cells to promote angiogenesis (43). Therefore, the observed reduction in Tregs, Th17 cells, and MDSCs in response to the MMPI in our study may explain the reduction in the MVD. Reducing the MVD through application of the MMPI could also further inhibit immune cell infiltration.

In a recent study in mice, inhibition of MMP-14 improved tumor blood perfusion and oxygen supply by increasing the expression of inducible nitric oxide synthase (iNOS) and heat-shock protein 90 (HSP90) and also resulted in bending of the blood vessels within the tumor (44). Vascular remodeling can improve the abnormal state of tumor blood vessels, enhance vascular perfusion, and increase drug delivery. In addition to MMP-14, MMP-2 plays an important role in promoting tumor vascular remodeling (45). Moreover, MMPI has been shown to enhance the therapeutic effects of chemotherapy drugs on tumors by promoting remodeling of the tumor vasculature and increasing drug delivery (46). Thus, the MMPI we used may promote tumor vascular remodeling to a certain extent.

CD4+ and CD8+ T cells are the main effector cells of antitumor immune responses and can combine with tumor associated antigens, MHC-I and MHC-II molecules to induce tumor cells death through secretion of cytokines and other cytotoxic effects. Thus, the number of CD4+ and CD8+ T cells can reflect the antitumor immunity of tumor-bearing mice. Unexpectedly, we did not see an increase in the number of CD4+ and CD8+ T cells after treatment. However, the ratio of CD8+ T cells to CD4+ T cells (CD8+/CD4+ T cells) was increased in tumor after combined treatment and in spleen after MMPI treatment. The different cell sources may be the cause for the different results. The characteristics of 4T1 cells themselves and the reduction in Tregs, a type of CD4+ T cell, may explain this unexpected result.

In conclusion, our data showed that the MMPI used in this study inhibited angiogenesis in the TME and suppressed the infiltration of immunosuppressive cells. Therefore, the MMPI may regulate blood vessels and immunosuppressive cells, together or separately, within the TME, thereby making the TME less favorable for tumor growth and enhancing the anticancer effects of anti-CTLA-4 antibody. Notably, tumor neovascularization and tumor infiltrating immune cells are two main components in TME. The main target of CTLA-4 antibody and MMPI is the immune cells and blood vessels, respectively. Thus, we only examined the two main factors most relevant to tumor progression in the TME. There may be other factors related to these processes that have not yet been identified. Further studies are needed to determine the functions of immunosuppressive cells regulating blood vessels in the TME.

Acknowledgments

We thank Professor Xuexun Fang (Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, Jilin University, China) for providing the MMPI potassium ferricyanide {K3[Fe(CN)6. This study was supported by the Jilin Provincial Science and Technology Projects (grant no. 20130102084JC) and the Frontier Interdisciplinary Program of Norman Bethune Health Science Center of Jilin University (grant no. 2013101005).

Abbreviations:

MMP

matrix metalloproteinase

MMPI

matrix metalloproteinase inhibitor

CTLA-4

cytotoxic T lymphocyte antigen-4

TME

tumor microenvironment

Tregs

regulatory T cells

MDSCs

myeloid-derived suppressor cells

MVD

microvessel density

ALT

alanine aminotransferase

AST

aspartate aminotransferase

IL

interleukin

BSA

bovine serum albumin

VEGF

vascular endothelial growth factor

TGF

transforming growth factor

STAT3

signal transducer and activator of transcription

PGE2

prostaglandin E2

References

1 

Lotem M, Merims S, Frank S, Ospovat I and Peretz T: Ctla-4 blockade: A new hope for the immunotherapy of malignant melanoma. Harefuah. 151:585–588. 6042012.In Hebrew.

2 

Prieto PA, Yang JC, Sherry RM, Hughes MS, Kammula US, White DE, Levy CL, Rosenberg SA and Phan GQ: CTLA-4 blockade with ipilimumab: Long-term follow-up of 177 patients with metastatic melanoma. Clin Cancer Res. 18:2039–2047. 2012. View Article : Google Scholar : PubMed/NCBI

3 

McAllister SS and Weinberg RA: The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 16:717–727. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Baginska J, Viry E, Paggetti J, Medves S, Berchem G, Moussay E and Janji B: The critical role of the tumor microenvironment in shaping natural killer cell-mediated anti-tumor immunity. Front Immunol. 4:4902013. View Article : Google Scholar

5 

Whiteside TL: Induced regulatory T cells in inhibitory microenvironments created by cancer. Expert Opin Biol Ther. 14:1411–1425. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Borriello L and DeClerck YA: Tumor microenvironment and therapeutic resistance process. Med Sci (Paris). 30:445–451. 2014.In French. View Article : Google Scholar

7 

Hida K, Akiyama K, Ohga N, Maishi N and Hida Y: Tumour endothelial cells acquire drug resistance in a tumour microenvironment. J Biochem. 153:243–249. 2013. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

Shuman Moss LA, Jensen-Taubman S and Stetler-Stevenson WG: Matrix metalloproteinases: Changing roles in tumor progression and metastasis. Am J Pathol. 181:1895–1899. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Thomas D, Ritz MF, Malviya AN and Gaillard S: Intracellular acidification mediates the proliferative response of PC12 cells induced by potassium ferricyanide and involves MAP kinase activation. Int J Cancer. 68:547–552. 1996. View Article : Google Scholar : PubMed/NCBI

11 

Martinus RD, Linnane AW and Nagley P: Growth of rho 0 human Namalwa cells lacking oxidative phosphorylation can be sustained by redox compounds potassium ferricyanide or coenzyme Q10 putatively acting through the plasma membrane oxidase. Biochem Mol Biol Int. 31:997–1005. 1993.PubMed/NCBI

12 

Stockmann C, Schadendorf D, Klose R and Helfrich I: The impact of the immune system on tumor: Angiogenesis and vascular remodeling. Front Oncol. 4:692014. View Article : Google Scholar : PubMed/NCBI

13 

Tao K, Fang M, Alroy J and Sahagian GG: Imagable 4T1 model for the study of late stage breast cancer. BMC Cancer. 8:2282008. View Article : Google Scholar : PubMed/NCBI

14 

Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP and Formenti SC: Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res. 11:728–734. 2005.PubMed/NCBI

15 

Fink K and Boratyński J: The role of metalloproteinases in modification of extracellular matrix in invasive tumor growth, metastasis and angiogenesis. Postepy Hig Med Dosw Online. 66:609–628. 2012.In Polish. View Article : Google Scholar

16 

Benson CS, Babu SD, Radhakrishna S, Selvamurugan N and Ravi Sankar B: Expression of matrix metalloproteinases in human breast cancer tissues. Dis Markers. 34:395–405. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Overall CM and Kleifeld O: Tumour microenvironment - opinion: Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nat Rev Cancer. 6:227–239. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Li NG, Tang YP, Duan JA and Shi ZH: Matrix metalloproteinase inhibitors: A patent review (2011–2013). Expert Opin Ther Pat. 24:1039–1052. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Li W, Saji S, Sato F, Noda M and Toi M: Potential clinical applications of matrix metalloproteinase inhibitors and their future prospects. Int J Biol Markers. 28:117–130. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Srivastava MK, Zhu L, Harris-White M, Huang M, St John M, Lee JM, Salgia R, Cameron RB, Strieter R, Dubinett S, et al: Targeting myeloid-derived suppressor cells augments antitumor activity against lung cancer. Immunotargets Ther. 2012:7–12. 2012.PubMed/NCBI

21 

Radosavljević GD, Jovanović IP, Kanjevac TV and Arsenijević NN: The role of regulatory T cells in the modulation of anti-tumor immune response. Srp Arh Celok Lek. 141:262–267. 2013.In Serbian. View Article : Google Scholar

22 

Prabhala RH, Pelluru D, Fulciniti M, Prabhala HK, Nanjappa P, Song W, Pai C, Amin S, Tai YT, Richardson PG, et al: Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood. 115:5385–5392. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Becker JC, Andersen MH, Schrama D and Thor Straten P: Immune-suppressive properties of the tumor microenvironment. Cancer Immunol Immunother. 62:1137–1148. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Walker LS: Treg and CTLA-4: Two intertwining pathways to immune tolerance. J Autoimmun. 45:49–57. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Nagaraj S, Youn JI and Gabrilovich DI: Reciprocal relationship between myeloid-derived suppressor cells and T cells. J Immunol. 191:17–23. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Li Z, Li D, Tsun A and Li B: FOXP3+ regulatory T cells and their functional regulation. Cell Mol Immunol. 12:558–565. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Kumar S, Pan CC, Bloodworth JC, Nixon AB, Theuer C, Hoyt DG and Lee NY: Antibody-directed coupling of endoglin and MMP-14 is a key mechanism for endoglin shedding and deregulation of TGF-β signaling. Oncogene. 33:3970–3979. 2014. View Article : Google Scholar :

28 

Krstic J and Santibanez JF: Transforming growth factor-beta and matrix metalloproteinases: Functional interactions in tumor stroma-infiltrating myeloid cells. Sci World J. 2014:5217542014. View Article : Google Scholar

29 

Guedez L, Jensen-Taubman S, Bourboulia D, Kwityn CJ, Wei B, Caterina J and Stetler-Stevenson WG: TIMP-2 targets tumor-associated myeloid suppressor cells with effects in cancer immune dysfunction and angiogenesis. J Immunother. 35:502–512. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Fan Q, Gu D, Liu H, Yang L, Zhang X, Yoder MC, Kaplan MH and Xie J: Defective TGF-β signaling in bone marrow-derived cells prevents hedgehog-induced skin tumors. Cancer Res. 74:471–483. 2014. View Article : Google Scholar :

31 

Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, et al: Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer. 124:2621–2633. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Hida K, Kawamoto T, Ohga N, Akiyama K, Hida Y and Shindoh M: Altered angiogenesis in the tumor microenvironment. Pathol Int. 61:630–637. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Goubran HA, Kotb RR, Stakiw J, Emara ME and Burnouf T: Regulation of tumor growth and metastasis: The role of tumor microenvironment. Cancer growth Metastasis. 7:9–18. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Mittal K, Ebos J and Rini B: Angiogenesis and the tumor microenvironment: Vascular endothelial growth factor and beyond. Semin Oncol. 41:235–251. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Chauhan VP, Stylianopoulos T, Martin JD, Popović Z, Chen O, Kamoun WS, Bawendi MG, Fukumura D and Jain RK: Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat Nanotechnol. 7:383–388. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Trédan O, Lacroix-Triki M, Guiu S, Mouret-Reynier MA, Barrière J, Bidard FC, Braccini AL, Mir O, Villanueva C and Barthélémy P: Angiogenesis and tumor microenvironment: Bevacizumab in the breast cancer model. Target Oncol. 10:189–198. 2015. View Article : Google Scholar

37 

Sounni NE, Paye A, Host L and Noël A: MT-MMPS as regulators of vessel stability associated with angiogenesis. Front Pharmacol. 2:1112011. View Article : Google Scholar : PubMed/NCBI

38 

Pucino V, De Rosa V, Procaccini C and Matarese G: Regulatory T cells, leptin and angiogenesis. Chem Immunol Allergy. 99:155–169. 2014. View Article : Google Scholar

39 

D'Alessio FR, Zhong Q, Jenkins J, Moldobaeva A and Wagner EM: Lung angiogenesis requires CD4 (+) forkhead homeobox protein-3 (+) regulatory T cells. Am J Respir Cell Mol Biol. 52:603–610. 2015. View Article : Google Scholar

40 

Mucha J, Majchrzak K, Taciak B, Hellmén E and Król M: MDSCs mediate angiogenesis and predispose canine mammary tumor cells for metastasis via IL-28/IL-28RA (IFN-λ) signaling. PLoS One. 9:e1032492014. View Article : Google Scholar

41 

Finke J, Ko J, Rini B, Rayman P, Ireland J and Cohen P: MDSC as a mechanism of tumor escape from sunitinib mediated anti-angiogenic therapy. Int Immunopharmacol. 11:856–861. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Yang B, Kang H, Fung A, Zhao H, Wang T and Ma D: The role of interleukin 17 in tumour proliferation, angiogenesis, and metastasis. Mediators Inflamm. 2014:6237592014. View Article : Google Scholar : PubMed/NCBI

43 

Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T, Robbins PD, Tahara H and Lotze MT: Interleukin-17 promotes angiogenesis and tumor growth. Blood. 101:2620–2627. 2003. View Article : Google Scholar

44 

Ager EI, Kozin SV, Kirkpatrick ND, Seano G, Kodack DP, Askoxylakis V, Huang Y, Goel S, Snuderl M, Muzikansky A, et al: Blockade of MMP14 activity in murine breast carcinomas: Implications for macrophages, vessels, and radiotherapy. J Natl Cancer Inst. 107:1072015. View Article : Google Scholar

45 

Romanchikova N, Trapencieris P, Zemītis J and Turks M: A novel matrix metalloproteinase-2 inhibitor triazolylmethyl aziridine reduces melanoma cell invasion, angiogenesis and targets ERK1/2 phosphorylation. J Enzyme Inhib Med Chem. 29:765–772. 2014. View Article : Google Scholar

46 

Weisshardt P, Trarbach T, Dürig J, Paul A, Reis H, Tilki D, Miroschnik I, Ergün S and Klein D: Tumor vessel stabilization and remodeling by anti-angiogenic therapy with bevacizumab. Histochem Cell Biol. 137:391–401. 2012. View Article : Google Scholar

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li M, Xing S, Zhang H, Shang S, Li X, Ren B, Li G, Chang X, Li Y, Li W, Li W, et al: A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment. Oncol Rep 35: 1329-1339, 2016
APA
Li, M., Xing, S., Zhang, H., Shang, S., Li, X., Ren, B. ... Li, W. (2016). A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment. Oncology Reports, 35, 1329-1339. https://doi.org/10.3892/or.2016.4547
MLA
Li, M., Xing, S., Zhang, H., Shang, S., Li, X., Ren, B., Li, G., Chang, X., Li, Y., Li, W."A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment". Oncology Reports 35.3 (2016): 1329-1339.
Chicago
Li, M., Xing, S., Zhang, H., Shang, S., Li, X., Ren, B., Li, G., Chang, X., Li, Y., Li, W."A matrix metalloproteinase inhibitor enhances anti-cytotoxic T lymphocyte antigen-4 antibody immunotherapy in breast cancer by reprogramming the tumor microenvironment". Oncology Reports 35, no. 3 (2016): 1329-1339. https://doi.org/10.3892/or.2016.4547