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.
Vaccine ; 36(25): 3708-3716, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29752021

ABSTRACT

Most active cancer immunotherapies able to induce a long-lasting protection against tumours are based on the activation of tumour-specific cytotoxic T lymphocytes (CTLs). Cell death by hyperthermia induces apoptosis followed by secondary necrosis, with the production of factors named "danger associated molecular pattern" (DAMP) molecules (DAMPs), that activate dendritic cells (DCs) to perform antigen uptake, processing and presentation, followed by CTLs cross priming. In many published studies, hyperthermia treatment of tumour cells is performed at 42-45 °C; these temperatures mainly promote cell surface expression of DAMPs. Treatment at 56 °C of tumour cells was shown to induce DAMPs secretion rather than their cell surface expression, improving DC activation and CTL cross priming in vitro. Thus we tested the relevance of this finding in vivo on the generation of a tumour-specific memory immune response, in the TRAMP-C2 mouse prostate carcinoma transplantable model. TRAMP-C2 tumour cells treated at 56 °C were able not only to activate DCs in vitro but also to trigger a tumour-specific CTL-dependent immune response in vivo. Prophylactic vaccination with 56 °C-treated TRAMP-C2 tumour cells alone provided protection against TRAMP-C2 tumour growth in vivo, whilst in the therapeutic regimen, control of tumour growth was achieved combining immunization with adjuvant chemotherapy.


Subject(s)
Combined Modality Therapy/methods , Dendritic Cells/immunology , Epithelial Cells/transplantation , Hyperthermia, Induced/methods , Immunotherapy/methods , Prostatic Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antineoplastic Agents/pharmacology , Chemotherapy, Adjuvant/methods , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Prostate/drug effects , Prostate/immunology , Prostate/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/pathology , Tumor Burden/drug effects , Tumor Cells, Cultured
2.
Immunology ; 146(1): 33-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25959091

ABSTRACT

In the Sp6 mouse plasmacytoma model, a whole-cell vaccination with Sp6 cells expressing de novo B7-1 (Sp6/B7) induced anatomically localized and cytotoxic T cell (CTL)-mediated protection against wild-type (WT) Sp6. Both WT Sp6 and Sp6/B7 showed down-regulated expression of MHC H-2 L(d). Increase of H-2 L(d) expression by cDNA transfection (Sp6/B7/L(d)) raised tumour immune protection and shifted most CTL responses towards H-2 L(d)-restricted antigenic epitopes. The tumour-protective responses were not specific for the H-2 L(d)-restricted immunodominant AH1 epitope of the gp70 common mouse tumour antigen, although WT Sp6 and transfectants were able to present it to specific T cells in vitro. Gp70 transcripts, absent in secondary lymphoid organs of naive mice, were detected in immunized mice as well as in splenocytes from naive mice incubated in vitro with supernatants of CTL-lysed Sp6 cell cultures, containing damage-associated molecular patterns (DAMPs). It has been shown that Toll-like receptor triggering induces gp70 expression. Damage-associated molecular patterns are released by CTL-mediated killing of Sp6/B7-Sp6/B7/L(d) cells migrated to draining lymph nodes during immunization and may activate gp70 expression and presentation in most resident antigen-presenting cells. The same could also apply for Mus musculus endogenous ecotropic murine leukaemia virus 1 particles present in Sp6-cytosol, discharged by dying cells and superinfecting antigen-presenting cells. The outcome of such a massive gp70 cross-presentation would probably be tolerogenic for the high-affinity AH1-gp70-specific CTL clones. In this scenario, autologous whole-tumour-cell vaccines rescue tumour-specific immunoprotection by amplification of subdominant tumour antigen responses when those against the immune dominant antigens are lost.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , H-2 Antigens/immunology , Plasmacytoma/immunology , Animals , Antigens, Neoplasm/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Movement/immunology , Female , H-2 Antigens/biosynthesis , H-2 Antigens/genetics , HMGB1 Protein/metabolism , Leukemia Virus, Murine/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Depletion , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred BALB C , Plasmacytoma/therapy , T-Lymphocytes, Cytotoxic/immunology , Vaccination
3.
Stem Cells Dev ; 20(4): 709-19, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20695752

ABSTRACT

Bone marrow mesenchymal stromal cells (BM-MSCs) may survive and proliferate in the presence of cycling neoplastic cells. Exogenously administered MSCs are actively incorporated in the tumor as stromal fibroblasts, thus competing with the local mesenchymal cell precursors. For this reason, MSCs have been suggested as a suitable carrier for gene therapy strategies, as they can be genetically engineered with genes encoding for biologically active molecules that can inhibit tumor cell proliferation and enhance the antitumor immune response. We used BM-MSCs engineered with the murine interferon-alpha (IFN-α) gene (BM-MSCs/IFN-α) to assess in a mouse plasmacytoma model the efficacy of this approach toward neoplastic plasma cells. We found that IFN-α can be efficiently produced and delivered inside the tumor microenvironment. Subcutaneous multiple administration of BM-MSCs/IFN-α significantly hampered the tumor growth in vivo and prolonged the overall survival of mice. The antitumor effect was associated with enhanced apoptosis of tumor cells, reduction in microvessel density, and ischemic necrosis. By contrast, intravenous administration of BM-MSCs/IFN-α did not significantly modify the survival of mice, mainly as a consequence of an excessive entrapment of injected cells in the pulmonary vessels. In conclusion, BM-MSCs/IFN-α are effective in inhibiting neoplastic plasma cell growth; however, systemic administration of engineered MSCs needs to be improved to make this approach potentially suitable for the treatment of multiple myeloma.


Subject(s)
Interferon-alpha/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Plasmacytoma/therapy , Animals , Apoptosis , Bone Marrow Cells/cytology , Cell Line, Tumor , Cell Survival , Coculture Techniques , Genetic Therapy , Interferon-alpha/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neovascularization, Pathologic/therapy , Plasma Cells/pathology , Plasmacytoma/blood supply , Plasmacytoma/pathology , Recombinant Proteins/metabolism , Thy-1 Antigens/metabolism , Transplantation, Heterologous , Tumor Burden
4.
Vaccine ; 28(20): 3548-57, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20304037

ABSTRACT

De novo expression of B7-1 impaired tumorigenicity of TRAMP-C2 mouse prostate adenocarcinoma (TRAMP-C2/B7), but it did not elicit a protective response against TRAMP-C2 parental tumor, unless after in vitro treatment with IFN-gamma. TRAMP-C2 cells secrete TGF-beta and show low MHC-I expression. Treatment with IFN-gamma increased MHC-I expression by induction of some APM components and antagonizing the immunosuppressant activity of TGF-beta. Thus, immunization with TRAMP-C2/B7 conferred protection against TRAMP-C2-derived tumors in function of the IFN-gamma-mediated fine-tuned modulation of either APM expression or TGF-beta signaling. To explore possible clinical translation, we delivered IFN-gamma to TRAMP-C2 tumor site by means of genetically engineered MSCs secreting IFN-gamma.


Subject(s)
Adenocarcinoma/immunology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Prostatic Neoplasms/immunology , Adenocarcinoma/genetics , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, MHC Class I , Histocompatibility Antigens Class I/genetics , Male , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Prostatic Neoplasms/genetics , Spleen/cytology , Spleen/immunology , Transfection , Transforming Growth Factor beta/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...