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.
J Immunol ; 173(9): 5458-66, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494493

ABSTRACT

Dendritic cell (DC) migration is crucial for the initiation of immune responses. The balance between metalloproteinases (MMP) and tissue inhibitors of metalloproteinases (TIMP) has been shown to modulate DC migration. PGE2, which is overproduced in a wide variety of human malignancies, has been implicated in MMP and TIMP regulation in various cells, including monocytes. In the present study, we hypothesized that tumor-derived PGE2 would affect DC migratory capacity through the extracellular matrix (ECM) by altering MMP and TIMP balance. Treatment of monocyte-derived immature DC with exogenous PGE2 induced TIMP-1 secretion but not MMP-9 production and was correlated with reduced DC migration through ECM. Because recombinant TIMP-1 replicated PGE2 inhibition of DC migration while anti-TIMP-1 neutralizing Ab reversed it, we conclude that PGE2-mediated induction of TIMP-1 was responsible for the reduced migration of PGE2-treated DC. Similarly, DC cultured for 48 h in supernatants from cyclooxygenase-2 overexpressing lung cancer cells that secrete high levels of PGE2, exhibited decreased migration through ECM. Finally, analysis of E prostanoid receptor expression and their selective inhibition revealed that the enhanced TIMP-1 secretion in PGE2-treated DC was mediated predominantly by the E prostanoid receptor 2. These findings indicate that PGE2-dependent enhancement of TIMP-1 production causes reduced migration of DC through ECM.


Subject(s)
Adjuvants, Immunologic/physiology , Cell Migration Inhibition , Dendritic Cells/cytology , Dendritic Cells/enzymology , Dinoprostone/physiology , Extracellular Matrix/enzymology , Extracellular Matrix/immunology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , 16,16-Dimethylprostaglandin E2/metabolism , 16,16-Dimethylprostaglandin E2/pharmacology , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Immunologic , Humans , Matrix Metalloproteinase 9/metabolism , Receptors, CCR7 , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/biosynthesis , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP2 Subtype , Signal Transduction/immunology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transfection , Up-Regulation/immunology
2.
Cancer Res ; 64(18): 6549-55, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374967

ABSTRACT

Constitutive overexpression of cyclooxygenase-2 (COX-2) occurs frequently in several different malignancies, including lung, colon, breast, and prostate cancer. Clinical studies have established elevated serum insulin-like growth factor (IGF-I) content and IGF-I:IGF-binding protein 3 (IGFBP-3) ratio as risk factors for these same malignancies. Therefore, we sought to determine the link between COX-2 expression and the IGF axis in COX-2 gene-modified human non-small-cell lung cancer (NSCLC) cells. Overexpression of COX-2 in NSCLC cells enhanced the antiapoptotic and mitogenic effects of IGF-I and IGF-II, facilitated the autophosphorylation of the type 1 IGF receptor, increased class IA phosphatidylinositol 3'-kinase activity, and decreased expression of IGFBP-3. Thus, these findings show that COX-2 augments the stimulatory arm of the IGF axis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Isoenzymes/physiology , Lung Neoplasms/enzymology , Prostaglandin-Endoperoxide Synthases/physiology , Receptor, IGF Type 1/metabolism , Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Celecoxib , Cell Division/physiology , Cell Line, Tumor , Cell Survival/physiology , Cyclooxygenase 2 , DNA, Antisense/genetics , Down-Regulation/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Pyrazoles , Signal Transduction/physiology , Sulfonamides/pharmacology
3.
Drug Resist Updat ; 7(3): 169-84, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15296859

ABSTRACT

Lung cancer is the leading cause of cancer death in the United States. Although the low 5-year survival rate (under 15%) has changed minimally in the last 25 years, new agents and combinations of agents that target tumor proliferation, invasion, and survival may lead to improvement in patient outcomes. There is evidence that cyclooxygenase-2 (COX-2) is overexpressed in lung cancer and promotes tumor proliferation, invasion, angiogenesis, and resistance to apoptosis. COX-2 inhibitors have been found to inhibit tumor growth in animal models and have demonstrated responses when combined with conventional therapy in phase II clinical trials. Further understanding of the mechanisms involved in COX-2-mediated tumorigenesis and its interaction with other molecules in lung cancer may lead to improved therapeutic strategies for this disease. In addition, delineation of how COX-2-dependent genes modulate the malignant phenotype will provide novel insights in lung cancer pathogenesis.


Subject(s)
Isoenzymes/physiology , Lung Neoplasms/etiology , Prostaglandin-Endoperoxide Synthases/physiology , Antigen-Presenting Cells/physiology , Apoptosis , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/physiology , ErbB Receptors/antagonists & inhibitors , Humans , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Membrane Proteins , Neovascularization, Pathologic/etiology
4.
Clin Cancer Res ; 10(8): 2891-901, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15102698

ABSTRACT

To achieve in situ tumor antigen uptake and presentation, intratumoral administration of ex vivo-generated, gene-modified murine bone marrow-derived dendritic cells (DC) was used in a murine lung cancer model. To attract mature host DC and activated T cells at the tumor site, the DC were transduced with an adenoviral vector expressing secondary lymphoid tissue chemokine (CCL21/SLC). Sixty percent of the mice treated with 10(6) DC-AdCCL21 intratumorally (7-10 ng/ml/10(6) cells/24 h of CCL21) at weekly intervals for 3 weeks showed complete tumor eradication, whereas only 25% of mice had complete resolution of tumors when mice were treated with fibroblasts expressing CCL21. In contrast only 12% of the mice treated with unmodified or control vector modified DC (DC-AdCV) showed complete tumor eradication. DC-AdCCL21 administration led to increases in the CD4(+), CD8(+), and CD3(+)CXCR3(+) T cells, as well as DC expressing CD11c(+) DEC205(+). CD4(+)CD25(+) T-regulatory cells infiltrating the tumors were markedly reduced after DC-AdCCL21 therapy. The tumor site cellular infiltrates were accompanied by the enhanced elaboration of granulocyte macrophage colony-stimulating factor, IFN-gamma, MIG/CXCL9, IP-10/CXCL10, and interleukin 12, but decreases in the immunosuppressive mediators transforming growth factor beta and prostaglandin E(2). DC-AdCCL21-treated tumor-bearing mice showed enhanced frequency of tumor-specific T lymphocytes secreting IFN-gamma, and tumor protective immunity was induced after DC-AdCCL21 therapy. In vivo depletion of IP-10/CXCL10, MIG/CXCL9, or IFN-gamma significantly reduced the antitumor efficacy of DC-AdCCL21. These findings provide a strong rationale for the evaluation of DC-AdCCL21 in cancer immunotherapy.


Subject(s)
Cancer Vaccines , Dendritic Cells/immunology , Immunotherapy/methods , Adenoviridae/genetics , Animals , Antineoplastic Agents/pharmacology , CD3 Complex/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CCL21 , Chemokines, CC/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunoenzyme Techniques , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , T-Lymphocytes/metabolism , Time Factors , Transforming Growth Factor beta/metabolism
5.
Mol Cancer ; 2: 35, 2003 Nov 02.
Article in English | MEDLINE | ID: mdl-14613584

ABSTRACT

BACKGROUND: Ex vivo generated dendritic cells (DC) genetically modified to express secondary lymphoid tissue chemokine (CCL-21/SLC) have been shown to stimulate potent antitumor responses in murine models. When injected intratumorally, CCL-21 colocalizes DC and lymphocyte effector cells at the tumor site. This may improve tumor antigen presentation and T cell activation by utilizing the tumor as an in vivo source of antigen for DC. In order to develop DC-based cancer therapies for intratumoral injection that could promote tumor antigen uptake and presentation in situ, we constructed and characterized an adenoviral vector that expresses human CCL-21 (AdCCL-21). RESULTS: Human monocyte derived DC were cultured in GM-CSF and IL-4 for 6 days. Following AdCCL-21 transduction, CCL-21 protein production was assessed by ELISA on day 8. DC transduced with AdCCL-21 at multiplicities of infection (MOIs) of 50:1 or 100:1 produced up to 210 +/- 9 ng/ml and 278 +/- 6.5 ng/ml /106 cells/48 hours, respectively. Following transduction, an immature DC phenotype was maintained and an upregulation of the costimulatory molecule, CD86 was noted. In addition, supernatant from AdCCL-21-DC caused significant chemotaxis of peripheral blood lymphocytes and mature DC. CONCLUSIONS: These studies demonstrate that AdCCL-21-DC generate functional levels of CCL-21 without adversely altering DC phenotype. These findings strengthen the rationale for further investigation of AdCCL-21-DC as a DC-based therapy in cancer treatment.


Subject(s)
Antigen-Presenting Cells/physiology , Chemokines, CC/metabolism , Dendritic Cells/metabolism , Lymphocytes/physiology , Adenoviridae/genetics , Antibodies, Monoclonal/pharmacology , Antigen-Presenting Cells/cytology , Cell Movement/drug effects , Chemokine CCL21 , Chemokines, CC/genetics , Chemokines, CC/immunology , Chemotaxis/drug effects , Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Flow Cytometry , Genetic Vectors/genetics , Humans , Immunophenotyping , Interferon-gamma/pharmacology , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Transfection
6.
Hum Gene Ther ; 14(16): 1511-24, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14577913

ABSTRACT

The antitumor efficiency of dendritic cells transduced with an adenovirus vector expressing interleukin (IL)-7 (DC-AdIL-7) was evaluated in a murine model of spontaneous bronchoalveolar cell carcinoma. These transgenic mice (CC-10 TAg), expressing the SV40 large T antigen under the Clara cell promoter, develop bilateral multifocal pulmonary adenocarcinomas and die at 4 months as a result of progressive pulmonary tumor burden. Injection of DC-AdIL-7 in the axillary lymph node region (ALNR) weekly for 3 weeks led to a marked reduction in tumor burden with extensive lymphocytic infiltration of the tumors and enhanced survival. The antitumor responses were accompanied by the enhanced elaboration of interferon (IFN)-gamma and IL-12 as well as an increase in the antiangiogenic chemokines, IFN-gamma-inducible protein 10 (IP-10/CXCL10) and monokine induced by IFN-gamma (MIG/CXCL9). In contrast, production of the immunosuppressive mediators IL-10, transforming growth factor (TGF)-beta, prostaglandin E(2) (PGE(2)), and the proangiogenic modulator vascular endothelial growth factor (VEGF) decreased in response to DC-AdIL-7 treatment. Significant reduction in tumor burden in a model in which tumors develop in an organ-specific manner provides a strong rationale for further evaluation of DC-AdIL-7 in regulation of tumor immunity and its use in lung cancer genetic immunotherapy.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/immunology , Adenocarcinoma, Bronchiolo-Alveolar/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive , Interleukin-7/genetics , Lung Neoplasms/therapy , Adenoviridae/genetics , Animals , Genetic Vectors , Lung Neoplasms/immunology , Lymph Nodes/immunology , Mice , Mice, Transgenic , Remission Induction , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...