Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Oncol Rep ; 28(2): 707-13, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22581112

ABSTRACT

Many cancers express an array of chemokines which have the capacity to modulate the nature and function of intratumoural leukocyte infiltrates. In malignant mesothelioma (MM) neither the chemokine signalling networks nor their regulation have been investigated despite the prominence of leucocytic infiltrates in both clinical and experimental tumours. In this study, we examined constitutive and cytokine-regulated expression of CC and CXC chemokine genes in mesothelioma and mesothelial cell cultures derived from two different mouse strains (BALB/C and CBA/CaH). In mouse MM and mesothelial cells MCP-1/JE, GRO-α/KC and RANTES were expressed whereas MIP-1α and MIP-2 were infrequently expressed. Comparison of basal chemokine expression showed that GRO-α/KC mRNA was overexpressed in the malignant cells whereas MCP-1 gene expression and release was downregulated. Treatment of mesothelioma cells with IL-4, IFN-γ or TNF-α revealed that chemokine genes could be more responsive to cytokines in the malignant compared to their mesothelial cells. TNF-α was consistently the most potent positive regulator of both CC and CXC chemokine expression and MCP-1 release. The present study for the first time provides a mechanistic insight into the differential regulation of chemokine expression in malignant mesothelioma cells and has implications for mesothelial chemokine signalling in mouse models.


Subject(s)
Chemokines, CC/genetics , Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Mesothelioma/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Line, Tumor , Chemokines, CC/biosynthesis , Chemokines, CC/metabolism , Chemokines, CXC/biosynthesis , Chemokines, CXC/metabolism , Disease Models, Animal , Female , Mesothelioma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
2.
PLoS One ; 4(9): e6982, 2009 Sep 10.
Article in English | MEDLINE | ID: mdl-19746156

ABSTRACT

BACKGROUND: Anti-cancer chemotherapy can be simultaneously lymphodepleting and immunostimulatory. Pre-clinical models clearly demonstrate that chemotherapy can synergize with immunotherapy, raising the question how the immune system can be mobilized to generate anti-tumor immune responses in the context of chemotherapy. METHODS AND FINDINGS: We used a mouse model of malignant mesothelioma, AB1-HA, to investigate T cell-dependent tumor resolution after chemotherapy. Established AB1-HA tumors were cured by a single dose of cyclophosphamide in a CD8 T cell- and NK cell-dependent manner. This treatment was associated with an IFN-alpha/beta response and a profound negative impact on the anti-tumor and total CD8 T cell responses. Despite this negative effect, CD8 T cells were essential for curative responses. The important effector molecules used by the anti-tumor immune response included IFN-gamma and TRAIL. The importance of TRAIL was supported by experiments in nude mice where the lack of functional T cells could be compensated by agonistic anti-TRAIL-receptor (DR5) antibodies. CONCLUSION: The data support a model in which chemotherapy sensitizes tumor cells for T cell-, and possibly NK cell-, mediated apoptosis. A key role of tumor cell sensitization to immune attack is supported by the role of TRAIL in tumor resolution and explains the paradox of successful CD8 T cell-dependent anti-tumor responses in the absence of CD8 T cell expansion.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/pharmacology , Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Female , Immunotherapy/methods , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology
3.
J Immunol ; 182(9): 5217-24, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380767

ABSTRACT

Topical application of tumors with the TLR7 agonist imiquimod is an effective adjunct treatment for a range of primary dermatological cancers. However, for therapy to be effective against a broad range of solid tumor types, it must promote a strong systemic antitumor response that targets metastases in addition to primary tumor. We therefore investigated the potential of locally delivered imiquimod to stimulate an effective systemic antitumor response in a murine model of malignant mesothelioma (AB1-HA) with primary and distal tumors (dual tumor). Persistent delivery of imiquimod into primary tumor significantly retarded tumor growth in all treated mice compared with vehicle control. This local antitumor immune response required both CD8 T cells and NK cells, but not CD4 T cells, and was reliant on type I IFN induction. In vivo CTL studies and Ly6A/E staining of lymphocytes suggested that local imiquimod treatment had indeed induced a systemic, Ag-specific CD8 response. However, notably this response was not sufficient to retard the growth of an untreated distal tumor. Because local imiquimod treatment did not induce significant CD4 T cell responses, we investigated the efficacy of combining imiquimod with agonistic CD40 Ab (as a surrogate for CD4 T cell help). Combination of locally delivered imiquimod with systemic anti-CD40 immunotherapy not only significantly enhanced the local antitumor response, with 30% complete resolution, but it was also effective at significantly retarding growth of distal tumor. These results demonstrate that antitumor responses induced by locally delivered TLR7 agonists can be harnessed systemically for treating distal tumor.


Subject(s)
Aminoquinolines/administration & dosage , Antibodies, Monoclonal/administration & dosage , CD40 Antigens/immunology , Membrane Glycoproteins/agonists , Mesothelioma/immunology , Mesothelioma/therapy , Toll-Like Receptor 7/agonists , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Aminoquinolines/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Therapy, Combination , Female , Imiquimod , Interferon Type I/administration & dosage , Interferon-gamma/administration & dosage , Killer Cells, Natural/immunology , Ligands , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Mesothelioma/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mice, Transgenic , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/therapeutic use
4.
Cancer Immunol Immunother ; 58(8): 1219-28, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19052741

ABSTRACT

Tumor cell death potentially engages with the immune system. However, the efficacy of anti-tumor chemotherapy may be limited by tumor-driven immunosuppression, e.g., through CD25+ regulatory T cells. We addressed this question in a mouse model of mesothelioma by depleting or reconstituting CD25+ regulatory T cells in combination with two different chemotherapeutic drugs. We found that the efficacy of cyclophosphamide to eradicate established tumors, which has been linked to regulatory T cell depletion, was negated by adoptive transfer of CD25+ regulatory T cells. Analysis of post-chemotherapy regulatory T cell populations revealed that cyclophosphamide depleted cycling (Ki-67(hi)) T cells, including foxp3+ regulatory CD4+ T cells. Ki-67(hi) CD4+ T cells expressed increased levels of two markers, TNFR2 and ICOS, that have been associated with a maximally suppressive phenotype according to recently published studies. This suggest that cyclophosphamide depletes a population of maximally suppressive regulatory T cells, which may explain its superior anti-tumor efficacy in our model. Our data suggest that regulatory T cell depletion could be used to improve the efficacy of anti-cancer chemotherapy regimens. Indeed, we observed that the drug gemcitabine, which does not deplete cycling regulatory T cells, eradicates established tumors in mice only when CD25+ CD4+ T cells are concurrently depleted. Cyclophosphamide could be used to achieve regulatory T cell depletion in combination with chemotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Immunosuppressive Agents/therapeutic use , Lymphocyte Depletion , Mesothelioma/drug therapy , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocytes, Regulatory/drug effects , Adoptive Transfer , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Inducible T-Cell Co-Stimulator Protein , Kaplan-Meier Estimate , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Mesothelioma/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes, Regulatory/immunology , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...