Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Crit Care Med ; 198(6): 777-787, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29617574

ABSTRACT

RATIONALE: Myeloid-derived suppressor cell (MDSC) expansion has been found to play a role in disease progression in patients with cancer. However, the characteristics of MDSCs in lung cancer are poorly understood. OBJECTIVES: We prospectively investigated MDSCs and inflammatory factors in tumor and peripheral blood samples from patients with resectable non-small cell lung cancer and studied their correlations with the disease prognosis. METHODS: A complex analysis of MDSC subsets and inflammatory mediators was performed using flow cytometry and a Bio-Plex assay. MEASUREMENTS AND MAIN RESULTS: A significant increase in the frequency of circulating monocytic (M)-MDSCs was observed in the patients with non-small cell lung cancer compared with the healthy donors (HDs). Moreover, the frequencies of M- and polymorphonuclear (PMN)-MDSCs were higher in tumors than in the peripheral blood of the same patients. This accumulation was associated with elevated concentrations of inflammatory mediators involved in MDSC migration to and activation in the tumor microenvironment. An analysis of the MDSC immunosuppressive pattern showed increased programmed death-ligand 1 expression on circulating cells from patients compared with HDs. Tumor PMN-MDSCs displayed higher programmed death-ligand 1 expression levels than the same cells in the peripheral blood. The frequency of CCR5 (C-C chemokine receptor 5) expression on circulating M-MDSCs was significantly higher in the patients than in the HDs. Clinical data analysis revealed negative correlations between recurrence-free survival and the frequencies of PMN-MDSCs and CCR5+ M-MDSCs in the circulation but not in tumors. CONCLUSIONS: Our findings suggest that the level of MDSCs in the peripheral blood but not in tumor tissues predicts recurrence after surgery.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Myeloid-Derived Suppressor Cells/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Flow Cytometry , Humans , Male , Middle Aged , Prospective Studies , Young Adult
2.
Cancer Res ; 78(1): 157-167, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29089297

ABSTRACT

Accumulation of myeloid-derived suppressor cells (MDSC) in melanoma microenvironment is supported by chemokine receptor/chemokine signaling. Although different chemokines were suggested to be involved in this process, the role of CCR5 and its ligands is not established. Using a Ret transgenic mouse melanoma model, we found an accumulation of CCR5+ MDSCs in melanoma lesions associated with both increased concentrations of CCR5 ligands and tumor progression. Tumor-infiltrating CCR5+ MDSCs displayed higher immunosuppressive activity than their CCR5- counterparts. Upregulation of CCR5 expression on CD11b+Gr1+ myeloid cells was induced in vitro by CCR5 ligands and other inflammatory factors. In melanoma patients, CCR5+ MDSCs were enriched at the tumor site and correlated with enhanced production of CCR5 ligands. Moreover, they exhibited a stronger immunosuppressive pattern compared with CCR5- MDSCs. Blocking CCR5/CCR5 ligand interactions increased survival of tumor-bearing mice and was associated with reduced migration and immunosuppressive potential of MDSCs in tumor lesions. Our findings define a critical role for CCR5 in recruitment and activation of MDSCs, suggesting a novel strategy for melanoma treatment.Significance: These findings validate the importance of the CCR5/CCR5 ligand axis not only for MDSC recruitment but also for further activation of their immunosuppressive functions in the tumor microenvironment, with potentially broad therapeutic implications, given existing clinically available inhibitors of this axis. Cancer Res; 78(1); 157-67. ©2017 AACR.


Subject(s)
Melanoma/immunology , Myeloid-Derived Suppressor Cells/pathology , Receptors, CCR5/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Movement , Female , Humans , Male , Melanoma/pathology , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Proto-Oncogene Proteins c-ret/genetics , Tumor Microenvironment/immunology
3.
Cell Rep ; 21(8): 2212-2222, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166611

ABSTRACT

Cells of hematopoietic origin can be subdivided into cells of the lymphoid lineage and those of the myeloid lineage, among which are myeloid-derived suppressor cells (MDSCs). The MDSCs can be further divided into CD11b+Ly6G-Ly6Chi monocytic (Mo) MDSCs and CD11b+Ly6G+Ly6Clow polymorphonuclear (PMN) MDSCs. Both subtypes support tumor growth and suppress anti-tumor immunity. Their accumulation at the tumor site includes mobilization from the bone marrow to the blood followed by colonization at the tumor site. The present study examines the mechanism by which PMN-MDSCs are mobilized from the BM to the blood to later accumulate at the tumor site. We show that the chemokine receptor CCR5 is a key driver of this event. We also show that, beyond chemoattraction, the interaction between CCR5 and its ligands promotes the proliferation of CCR5+ PMN-MDSCs at the BM and, later, potentiates their immune-suppressive activities at the tumor site in part by inducing arginase-1.


Subject(s)
Bone Marrow/metabolism , Myeloid Cells/metabolism , Neoplasms/pathology , Receptors, CCR5/metabolism , Animals , Antigens, Ly/metabolism , Arginase/metabolism , Cell Proliferation/physiology , Humans , Immune Tolerance/physiology , Mice , Monocytes/immunology , Myeloid Cells/immunology , Neutrophils/metabolism , Receptors, Chemokine/metabolism
4.
Cancer Immunol Immunother ; 66(8): 1015-1023, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28382399

ABSTRACT

Malignant melanoma is characterized by the development of chronic inflammation in the tumor microenvironment, leading to the accumulation of myeloid-derived suppressor cells (MDSCs). Using ret transgenic mouse melanoma model, we found a significant migration of MDSCs expressing C-C chemokine receptor (CCR)5 into primary tumors and metastatic lymph nodes, which was correlated with tumor progression. An increased CCR5 expression on MDSCs was associated with elevated concentrations of CCR5 ligands in melanoma microenvironment. In vitro experiments showed that the upregulation of CCR5 expression on CD11b+Gr1+ immature myeloid cells was induced by CCR5 ligands, IL-6, GM-CSF, and other inflammatory factors. Furthermore, CCR5+ MDSCs infiltrating melanoma lesions displayed a stronger immunosuppressive pattern than their CCR5- counterparts. Targeting CCR5/CCR5 ligand signaling via a fusion protein mCCR5-Ig, which selectively binds and neutralizes all three CCR5 ligands, increased the survival of tumor-bearing mice. This was associated with a reduced migration and immunosuppressive potential of tumor MDSCs. In melanoma patients, circulating CCR5+ MDSCs were increased as compared to healthy donors. Like in melanoma-bearing mice, we observed an enrichment of these cells and CCR5 ligands in tumors as compared to the peripheral blood. Our findings define a critical role for CCR5 not only in the recruitment but also in the activation of MDSCs in tumor lesions, suggesting that novel strategies of melanoma treatment could be based on blocking CCR5/CCR5 ligand interactions.


Subject(s)
Immunotherapy/trends , Melanoma, Experimental/immunology , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Receptors, CCR5/metabolism , Skin Neoplasms/immunology , Animals , Cell Differentiation , Gene Expression Regulation, Neoplastic , Humans , Immunity, Cellular , Mice , Receptors, CCR5/genetics , Tumor Microenvironment , Melanoma, Cutaneous Malignant
5.
Semin Immunopathol ; 39(3): 295-305, 2017 04.
Article in English | MEDLINE | ID: mdl-27787613

ABSTRACT

Tumor progression is known to be supported by chronic inflammatory conditions developed in the tumor microenvironment. It is characterized by the long-term secretion of various inflammatory soluble factors (including cytokines, chemokines, growth factors, reactive oxygen and nitrogen species, prostaglandins, etc.) and strong leukocyte infiltration. Among leukocytes infiltrating tumors, myeloid-derived suppressor cells (MDSCs) represent one of the most important players mediating immunosuppression and supporting tumor escape. These cells can strongly inhibit antitumor immune reactions mediated by T cells and NK cells. Moreover, MDSCs are generated, recruited to the tumor site, and activated not only under the influence of soluble inflammatory mediators but also due to extracellular vesicles (EVs) secreted by tumor cells. EVs play a key role in the formation of MDSCs via the conversion of normal myeloid cells and altering the normal myelopoiesis. In addition, EVs help create a suitable microenvironment for the metastatic process.


Subject(s)
Immunologic Surveillance/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Tumor Escape/immunology , Animals , Cell Differentiation , Cell Movement/immunology , Cell-Derived Microparticles/metabolism , Cytokines/metabolism , Extracellular Vesicles/metabolism , Humans , Immunosuppression Therapy , Inflammation Mediators/metabolism , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Tumor Microenvironment/immunology
6.
Vaccines (Basel) ; 4(4)2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27827871

ABSTRACT

The immunosuppressive tumor microenvironment represents not only one of the key factors stimulating tumor progression but also a strong obstacle for efficient tumor immunotherapy. Immunosuppression was found to be associated with chronic inflammatory mediators including cytokines, chemokines and growth factors produced by cancer and stroma cells. Long-term intensive production of these factors induces the formation of myeloid-derived suppressor cells (MDSCs) representing one of the most important players mediating immunosuppression. Moreover, MDSCs could not only inhibit anti-tumor immune reactions but also directly stimulate tumor growth and metastasis. Therefore, understanding the mechanisms of their generation, expansion, recruitment and activation is required for the development of novel strategies for tumor immunotherapy.

7.
J Immunother ; 39(9): 343-354, 2016.
Article in English | MEDLINE | ID: mdl-27741089

ABSTRACT

The immunosuppressive microenvironment of solid tumors renders adoptively transferred T cells hypofunctional. However, adenoviral delivery of immunostimulatory cytokines IL2 and TNFα can significantly improve the efficacy of adoptive T-cell therapy. Using ret transgenic mice that spontaneously develop skin malignant melanoma, we analyzed the mechanism of action of adenoviruses coding for IL2 and TNFα in combination with adoptive transfer of TCR-transgenic TRP-2-specific T cells. Following T-cell therapy and intratumoral virus injection, a significant increase in antigen-experienced, tumor-reactive PD-1 CD8 T cells was seen in both cutaneous lesions and in metastatic lymph nodes. A reverse correlation between tumor weight and the number of tumor-reactive PD-1 tumor-infiltrating lymphocytes (TILs) was observed, suggesting that these T cells could target and kill tumor cells. It is interesting to note that, local expression of cytokines did not affect intratumoral levels of T-regulatory cells (Tregs), which had previously been associated with systemic IL2 therapy. Instead, Ad5-IL2 induced upregulation of IL2 receptor α-chain (CD25) on conventional CD4CD25Foxp3 cells, indicating that these CD4 T cells may contribute to CD8 T-cell activation and/or homing. Signs of therapy-induced resistance were also observed as the expression of PD-L1 on tumor-infiltrating granulocytic myeloid-derived suppressor cells was upregulated as a reaction to PD-1+ TILs. Finally, beneficial ratios between tumor-reactive PD-1 CD8 TILs and immunosuppressive cell subsets (Tregs and nitric oxide-producing myeloid-derived suppressor cells) were observed in primary and secondary tumor sites, indicating that local delivery of IL2 and TNFα coding adenoviruses can systemically modify the cellular composition of the tumor microenvironment in favor of adoptively transferred T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Melanoma/therapy , Skin Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Adenoviridae/genetics , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Cells, Cultured , Genetic Vectors/genetics , Immunomodulation , Interleukin-2/genetics , Interleukin-2/metabolism , Melanoma/genetics , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-ret/genetics , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tumor Burden , Tumor Microenvironment , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
J Clin Invest ; 126(3): 921-37, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26854930

ABSTRACT

Melanoma prognosis is dictated by tumor-infiltrating lymphocytes, the migratory and functional behavior of which is guided by chemokine or cytokine gradients. Here, we retrospectively analyzed the expression patterns of 9 homing receptors (CCR/CXCR) in naive and memory CD4+ and CD8+ T lymphocytes in 57 patients with metastatic melanoma (MMel) with various sites of metastases to evaluate whether T cell CCR/CXCR expression correlates with intratumoral accumulation, metastatic progression, and/or overall survival (OS). Homing receptor expression on lymphocytes strongly correlated with MMel dissemination. Loss of CCR6 or CXCR3, but not cutaneous lymphocyte antigen (CLA), on circulating T cell subsets was associated with skin or lymph node metastases, loss of CXCR4, CXCR5, and CCR9 corresponded with lung involvement, and a rise in CCR10 or CD103 was associated with widespread dissemination. High frequencies of CD8+CCR9+ naive T cells correlated with prolonged OS, while neutralizing the CCR9/CCL25 axis in mice stimulated tumor progression. The expansion of CLA-expressing effector memory CD8+ T cells in response to a single administration of CTLA4 blockade predicted disease control at 3 months in 47 patients with MMel. Thus, specific CCR/CXCR expression patterns on circulating T lymphocytes may guide potential diagnostic and therapeutic approaches.


Subject(s)
Biomarkers, Tumor/metabolism , Lung Neoplasms/metabolism , Melanoma/metabolism , Receptors, Chemokine/metabolism , Skin Neoplasms/metabolism , T-Lymphocytes/metabolism , Adult , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Case-Control Studies , Cell Line, Tumor , Female , Humans , Ipilimumab , Kaplan-Meier Estimate , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/secondary , Mice, Inbred C57BL , Middle Aged , Neoplasm Transplantation , Proportional Hazards Models , ROC Curve , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology
9.
Mediators Inflamm ; 2014: 808695, 2014.
Article in English | MEDLINE | ID: mdl-24623963

ABSTRACT

Chronic ethanol abuse is known to increase susceptibility to infections after injury, in part, by modification of macrophage function. Several intracellular signalling mechanisms are involved in the initiation of inflammatory responses, including the nuclear factor-κB (NF-κB) pathway. In this study, we investigated the systemic and hepatic effect of chronic ethanol feeding on in vivo activation of NF-κB in NF-κB(EGFP) reporter gene mice. Specifically, the study focused on Kupffer cell proinflammatory cytokines IL-6 and TNF-α and activation of NF-κB after chronic ethanol feeding followed by in vitro stimulation with lipopolysaccharide (LPS). We found that chronic ethanol upregulated NF-κB activation and increased hepatic and systemic proinflammatory cytokine levels. Similarly, LPS-stimulated IL-1 ß release from whole blood was significantly enhanced in ethanol-fed mice. However, LPS significantly increased IL-6 and TNF-α levels. These results demonstrate that chronic ethanol feeding can improve the responsiveness of macrophage LPS-stimulated IL-6 and TNF-α production and indicate that this effect may result from ethanol-induced alterations in intracellular signalling through NF-κB. Furthermore, LPS and TNF-α stimulated the gene expression of different inflammatory mediators, in part, in a NF-κB-dependent manner.


Subject(s)
Endotoxins/chemistry , Ethanol/chemistry , Gene Expression Regulation , Kupffer Cells/cytology , Leukocytes/cytology , NF-kappa B/metabolism , Alcohol Drinking , Animals , Genes, Reporter , Green Fluorescent Proteins/metabolism , Inflammation , Interleukin-6/metabolism , Lipopolysaccharides/chemistry , Liver/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...