Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 57(8): 1263-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18236040

ABSTRACT

Promising anti-tumor responses have been observed in the clinic using monoclonal antibodies (mAbs) that block immune checkpoints. One concern with these therapeutic agents remains the potential induction of immune breakthrough events (IBEs) resulting from the disruption of T cell homeostasis or the breaking of tolerance to self antigens. As an approach to maintaining anti-tumor responses but decreasing the likelihood of these events, the local expression of a mAb in combination with a GM-CSF-secreting cancer immunotherapy was evaluated. Using anti-cytotoxic T lymphocyte antigen (CTLA)-4 as a model antibody to test this hypothesis, tumor cell lines were generated that expressed the full-length mAb in addition to GM-CSF. Evaluation of these cell lines in two therapeutic tumor models revealed that local, cell-mediated delivery of anti-CTLA-4 from a GM-CSF-secreting tumor cell immunotherapy activated potent anti-tumor responses and prolonged overall survival at significantly lower serum mAb levels in the host. Furthermore, lowering the systemic exposure of the host to the immune modulatory mAb correlated with reduced evidence of systemic autoimmunity. This approach has broad utility for the delivery of mAbs or proteins locally from cellular immunotherapies to minimize IBEs while retaining the potent therapeutic effects of such combination treatments.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, CD/immunology , Autoimmunity/immunology , Immunotherapy , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Animals , Antibodies, Monoclonal/genetics , CTLA-4 Antigen , Cell Line, Tumor , Cloning, Molecular , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hybridomas/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
2.
Cancer Immunol Immunother ; 56(10): 1653-65, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17410360

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapies have demonstrated long-lasting, and specific anti-tumor immune responses in animal models. The studies reported here specifically evaluate two aspects of the immune response generated by such immunotherapies: the persistence of irradiated tumor cells at the immunization site, and the breadth of the immune response elicited to tumor associated antigens (TAA) derived from the immunotherapy. To further define the mechanism of GM-CSF-secreting cancer immunotherapies, immunohistochemistry studies were performed using the B16F10 melanoma tumor model. In contrast to previous reports, our data revealed that the irradiated tumor cells persisted and secreted high levels of GM-CSF at the injection site for more than 21 days. Furthermore, dense infiltrates of dendritic cells were observed only in mice treated with GM-CSF-secreting B16F10 cells, and not in mice treated with unmodified B16F10 cells with or without concurrent injection of rGM-CSF. In addition, histological studies also revealed enhanced neutrophil and CD4+ T cell infiltration, as well as the presence of apoptotic cells, at the injection site of mice treated with GM-CSF-secreting tumor cells. To evaluate the scope of the immune response generated by GM-CSF-secreting cancer immunotherapies, several related B16 melanoma tumor cell subclones that exist as a result of genetic drift in the original cell line were used to challenge mice previously immunized with GM-CSF-secreting B16F10 cells. These studies revealed that GM-CSF-secreting cancer immunotherapies elicit T cell responses that effectively control growth of related but antigenically distinct tumors. Taken together, these studies provide important new insights into the mechanism of action of this promising novel cancer immunotherapy.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , Dendritic Cells/immunology , Disease Models, Animal , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Recombinant Proteins , T-Lymphocytes/immunology
3.
Clin Immunol ; 123(2): 155-65, 2007 May.
Article in English | MEDLINE | ID: mdl-17320482

ABSTRACT

IL-7 is known for its role in lymphopoiesis and T-cell homeostasis. In addition, its capacity to augment the immune response to weak or low affinity antigens makes it an ideal candidate to evaluate in combination with a GM-CSF-secreting tumor cell immunotherapy, which has been shown to elicit broad humoral and cellular immune responses. The studies reported here show that IL-7, when combined with a GM-CSF-secreting tumor cell immunotherapy, significantly prolonged the survival of tumor-bearing mice. The enhanced anti-tumor protection correlated with an increased number of activated dendritic cells (DC) and T cells in lymphoid tissues, such as the draining lymph nodes (DLN) and spleen. Moreover, an increased number of activated effector T cells were found in the tumor microenvironment, correlating with a more potent systemic tumor-specific T-cell response than each monotherapy alone. Taken together, these studies demonstrate that IL-7 augments the anti-tumor response of a GM-CSF-secreting tumor cell immunotherapy in preclinical models.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-7/therapeutic use , Melanoma, Experimental/therapy , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , CD11c Antigen/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cell Count , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interferon-gamma/analysis , Interleukin-7/genetics , Interleukin-7/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/immunology , Lysosomal-Associated Membrane Protein 1/analysis , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Membrane Glycoproteins/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Survival Analysis , gp100 Melanoma Antigen
4.
Clin Cancer Res ; 12(22): 6808-16, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121902

ABSTRACT

PURPOSE: The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy in combination with vascular endothelial growth factor (VEGF) blockage in preclinical models. EXPERIMENTAL DESIGN: Survival and immune response were monitored in the B16 melanoma and the CT26 colon carcinoma models. VEGF blockade was achieved by using a recombinant adeno-associated virus vector expressing a soluble VEGF receptor consisting of selected domains of the VEGF receptors 1 and 2 (termed sVEGFR1/R2). Dendritic cell and tumor infiltrating lymphocyte activation status and numbers were evaluated by fluorescence-activated cell sorting analysis. Regulatory T cells were quantified by their CD4+CD25hi and CD4+FoxP3+ phenotype. RESULTS: The present study established that GM-CSF-secreting tumor cell immunotherapy with VEGF blockade significantly prolonged the survival of tumor-bearing mice. Enhanced anti-tumor protection correlated with an increased number of activated CD4+ and CD8+ tumor-infiltrating T cells and a pronounced decrease in the number of suppressive regulatory T cells residing in the tumor. Conversely, overexpression of VEGF from tumors resulted in elevated numbers of regulatory T cells in the tumor, suggesting a novel mechanism of VEGF-mediated immune suppression at the tumor site. CONCLUSION: GM-CSF-secreting cancer immunotherapy and VEGF blockade increases the i.t. ratio of effector to regulatory T cells to provide enhanced antitumor responses. This therapeutic combination may prove to be an effective strategy for the treatment of patients with cancer.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunotherapy/methods , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , T-Lymphocytes, Regulatory/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Apoptosis , Carcinoma/therapy , Cell Count , Colonic Neoplasms/therapy , Combined Modality Therapy , Dendritic Cells/cytology , Fas Ligand Protein/physiology , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Lymphocytes, Tumor-Infiltrating/cytology , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/mortality , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Survival Analysis , T-Lymphocytes, Regulatory/cytology , Treatment Outcome , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , fas Receptor/analysis
5.
Mol Ther ; 13(5): 956-66, 2006 May.
Article in English | MEDLINE | ID: mdl-16580881

ABSTRACT

The presence of the blood-brain barrier complicates drug delivery in the development of therapeutic agents for the treatment of glioblastoma multiforme (GBM). The use of local gene transfer in the brain has the potential to overcome this delivery barrier by allowing the expression of therapeutic agents directly at the tumor site. In this study, we describe the development of a recombinant adeno-associated (rAAV) serotype 8 vector that encodes an optimized soluble inhibitor, termed sVEGFR1/R2, of vascular endothelial growth factor (VEGF). VEGF is an angiogenic factor highly up-regulated in GBM tumor tissue and correlates with disease progression. In subcutaneous models of GBM, VEGF inhibition following rAAV-mediated gene transfer significantly reduces overall tumor volume and increases median survival time following a single administration of vector. Using orthotopic brain tumor models of GBM, we find that direct intracranial administration of the rAAV-sVEGFR1/R2 vector to the tumor site demonstrates anti-tumor efficacy at doses that are not efficacious following systemic delivery of the vector. We propose that rAAV-mediated gene transfer of a potent soluble VEGF inhibitor in the CNS represents an effective antiangiogenic treatment strategy for GBM.


Subject(s)
Central Nervous System/metabolism , Dependovirus/genetics , Gene Transfer Techniques , Glioblastoma/therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Cell Line , Cell Line, Tumor , Dependovirus/classification , Female , Genetic Therapy/methods , Genetic Vectors , Humans , Male , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasm Transplantation , Rats , Rats, Nude , Receptors, Vascular Endothelial Growth Factor/metabolism , Serotyping , Solubility , Transplantation, Heterologous
6.
Cancer Res ; 65(15): 6901-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061674

ABSTRACT

The presence of metastases in regional lymph nodes is a strong indicator of poor patient survival in many types of cancer. It has recently been shown that the lymphangiogenic growth factor, vascular endothelial growth factor-C (VEGF-C), and its receptor, VEGF receptor-3 (VEGFR3), may play a pivotal role in the promotion of metastasis to regional lymph nodes. In this study, human prostate and melanoma tumor models that preferentially metastasize to the lymph nodes following s.c. tumor cell implantation were established from lymph node metastases via in vivo selection. Melanoma tumor cell sublines established from lymph node metastasis express higher amounts of VEGF-C than the parental tumor cells. The inhibition of tumor-derived VEGF-C with a soluble VEGFR3 decoy receptor, sVEGFR3-Fc, expressed via a recombinant adeno-associated viral vector, potently blocks tumor-associated lymphangiogenesis and tumor metastasis to the lymph nodes, when the treatment was initiated before the tumor implantation. In addition, sVEGFR3-Fc serum levels required for efficient blockade of lymph node metastases are strictly dependent on the VEGF-C levels generated by the primary tumor. Recombinant adeno-associated virus-mediated gene transfer of sVEGFR3-Fc may represent a feasible therapeutic strategy for blockade of lymphogenous metastasis.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-3/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Cell Line, Tumor , Female , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-3/biosynthesis , Vascular Endothelial Growth Factor Receptor-3/blood
SELECTION OF CITATIONS
SEARCH DETAIL