Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 16(4): 1191-205, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145179

ABSTRACT

PURPOSE: Transforming growth factor beta (TGFbeta) is a pleiotropic cytokine that affects tumor growth, metastasis, stroma, and immune response. We investigated the therapeutic efficacy of anti-TGFbeta receptor II (TGFbeta RII) antibody in controlling metastasis and tumor growth as well as enhancing antitumor immunity in preclinical tumor models. EXPERIMENTAL DESIGN: We generated neutralizing antibodies to TGFbeta RII and assessed the antibody effects on cancer, stroma, and immune cells in vitro. The efficacy and mechanism of action of the antibody as monotherapy and in combination with chemotherapy in suppression of primary tumor growth and metastasis were evaluated in several tumor models. RESULTS: Anti-TGFbeta RII antibody blocked TGFbeta RII binding to TGFbeta 1, 2, and 3, and attenuated the TGFbeta-mediated activation of downstream Smad2 kinase, invasion of cancer cells, motility of endothelial and fibroblast cells, and induction of immunosuppressive cells. Treatment with the antibody significantly suppressed primary tumor growth and metastasis and enhanced natural killer and CTL activity in tumor-bearing mice. Immunohistochemistry analysis showed cancer cell apoptosis and massive necrosis, and increased tumor-infiltrating T effector cells and decreased tumor-infiltrating Gr-1+ myeloid cells in the antibody-treated tumors. Fluorescence-activated cell sorting analysis indicated the significant reduction of peripheral Gr-1+/CD11b+ myeloid cells in treated animals. Concomitant treatment with the cytotoxic agent cyclophosphamide resulted in a significantly increased antitumor efficacy against primary tumor growth and metastasis. CONCLUSIONS: These preclinical data provide a foundation to support using anti-TGFbeta RII antibody as a therapeutic agent for TGFbeta RII-dependent cancer with metastatic capacity.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/pharmacology , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/immunology , Receptors, Transforming Growth Factor beta/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Receptor, Transforming Growth Factor-beta Type II , Smad2 Protein/metabolism , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 13(10): 3068-78, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17505010

ABSTRACT

PURPOSE: The importance of cellular communication with the extracellular matrix in regulating cellular invasion is well established. Selective disruption of communication links between cells and the local microenvironment by specifically targeting non-cellular matrix-immobilized cryptic extracellular matrix epitopes may represent an effective new clinical approach to limit tumor-associated angiogenesis. Therefore, we sought to determine whether the HU177 cryptic collagen epitope plays a functional role in regulating angiogenesis in vivo. EXPERIMENTAL DESIGN: We examined the expression and characterized the HU177 cryptic collagen epitope in vitro and in vivo using immunohistochemistry and ELISA. We examined potential mechanisms by which this cryptic collagen epitope may regulate angiogenesis using in vitro cell adhesion, migration, proliferation, and biochemical assays. Finally, we examined the whether blocking cellular interactions with the HU177 cryptic epitope plays a role in angiogenesis and tumor growth in vivo using the chick embryo model. RESULTS: The HU177 cryptic epitope was selectively exposed within tumor blood vessel extracellular matrix, whereas little was associated with quiescent vessels. An antibody directed to this cryptic site selectively inhibited endothelial cell adhesion, migration, and proliferation on denatured collagen type IV and induced increased levels of cyclin-dependent kinase inhibitor p27(KIP1). Systemic administration of mAb HU177 inhibited cytokine- and tumor-induced angiogenesis in vivo. CONCLUSIONS: We provide evidence for a new functional cryptic regulatory element within collagen IV that regulates tumor angiogenesis. These findings suggest a novel and highly selective approach for regulating angiogenesis by targeting a non-cellular cryptic collagen epitope.


Subject(s)
Collagen Type IV/metabolism , Endothelium, Vascular/metabolism , Epitopes/metabolism , Extracellular Matrix/metabolism , Neovascularization, Pathologic/etiology , Animals , Antibodies, Monoclonal/pharmacology , Biological Assay , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Chick Embryo , Collagen Type IV/immunology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endothelium, Vascular/drug effects , Epitopes/immunology , Humans , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Up-Regulation
3.
Cancer Res ; 67(9): 4353-63, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483349

ABSTRACT

Angiogenesis and tumor metastasis depend on extracellular matrix (ECM) remodeling and subsequent cellular interactions with these modified proteins. An in-depth understanding of how both endothelial and tumor cells use matrix-immobilized cryptic ECM epitopes to regulate invasive cell behavior may lead to the development of novel strategies for the treatment of human tumors. However, little is known concerning the existence and the functional significance of cryptic laminin epitopes in regulating angiogenesis and tumor cell metastasis. Here, we report the isolation and characterization of a synthetic peptide that binds to a cryptic epitope in laminin. The STQ peptide selectively bound denatured and proteolyzed laminin but showed little interaction with native laminin. The cryptic laminin epitope recognized by this peptide was selectively exposed within malignant melanoma in vivo, whereas little if any was detected in normal mouse skin. Moreover, the STQ peptide selectively inhibited endothelial and tumor cell adhesion, migration, and proliferation in vitro and inhibited angiogenesis, tumor growth, and experimental metastasis in vivo. This inhibitory activity was associated with a selective up-regulation of the cyclin-dependent kinase inhibitor P27(KIP1) and induction of cellular senescence. These novel findings suggest the existence of functionally relevant cryptic laminin epitopes in vivo and that selective targeting of these laminin epitopes may represent an effective new strategy for the treatment of malignant tumors by affecting both the endothelial and tumor cell compartments.


Subject(s)
Epitopes/immunology , Extracellular Matrix/immunology , Laminin/immunology , Melanoma/blood supply , Melanoma/immunology , Animals , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Cell Movement/physiology , Chick Embryo , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epitopes/metabolism , Extracellular Matrix/metabolism , Humans , Laminin/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peptides/genetics , Peptides/immunology , Protein Binding , Protein Denaturation
4.
Am J Pathol ; 168(5): 1576-86, 2006 May.
Article in English | MEDLINE | ID: mdl-16651624

ABSTRACT

Metastasis from the primary tumor to distant sites involves an array of molecules that function in an integrated manner. Proteolytic remodeling and subsequent tumor cell interactions with the extracellular matrix regulate tumor invasion. In previous studies, we have identified a cryptic epitope (HUIV26) that is specifically exposed after alterations in the triple helical structure of type IV collagen. Exposure of this cryptic epitope plays a fundamental role in the regulation of angiogenesis in vivo. However, little is known concerning the ability of tumor cells to interact with this cryptic site or whether this site regulates tumor cell metastasis in vivo. In this regard, many of the same cellular processes that regulate angiogenesis also contribute to tumor metastasis. Here we provide evidence that tumor cells such as B16F10 melanoma interact with denatured collagen type IV in part by recognizing the HUIV26 cryptic site. Systemic administration of a HUIV26 monoclonal antibody inhibited experimental metastasis of B16F10 melanoma in vivo. Taken together, our findings suggest that tumor cell interactions with the HUIV26 cryptic epitope play an important role in regulating experimental metastasis and that this cryptic element may represent a therapeutic target for controlling the spread of tumor cells to distant sites.


Subject(s)
Antibodies, Monoclonal/immunology , Collagen/chemistry , Collagen/immunology , Epitopes/immunology , Lung Neoplasms/secondary , Neoplasm Metastasis/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Chick Embryo , Collagen Type IV/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Lung/pathology , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis/genetics , Neoplasm Metastasis/therapy
5.
Am J Pathol ; 166(3): 901-11, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15743801

ABSTRACT

Cellular interaction with the extracellular matrix is thought to be a critical event in controlling angiogenesis and tumor growth. In our previous studies, genetically distinct noncollagenous (NC) domains of type-IV collagen were shown to interact with integrin receptors expressed on the surface of endothelial cells. Moreover, these NC1 domains were shown to inhibit angiogenesis in vivo. Here, we provide evidence that a recombinant form of the alpha2(IV)NC1 domain of type-IV collagen could bind integrins alpha1beta1 and alphavbeta3 expressed on melanoma cells and inhibit tumor cell adhesion in a ligand-specific manner. Systemic administration of recombinant alpha2(IV)NC1 domain potently inhibited M21 melanoma tumor growth within full thickness human skin and exhibited a dose-dependent inhibition of tumor growth in nude mice. Interestingly, alpha2(IV)NC1 domain enhanced cellular senescence in tumor cells in vitro and in vivo. Taken together, these results suggest that recombinant alpha2(IV)NC1 domain is not only a potent anti-angiogenic reagent, but it also directly impacts tumor cell behavior. Thus, alpha2(IV)NC1 domain represents a potent inhibitor of tumor growth by impacting both endothelial and tumor cell compartments.


Subject(s)
Collagen Type IV/chemistry , Collagen Type IV/physiology , Extracellular Matrix/metabolism , Neoplasms/pathology , Animals , Apoptosis , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cellular Senescence , Chick Embryo , Dose-Response Relationship, Drug , Humans , Immunohistochemistry , Ligands , Melanoma/metabolism , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasm Transplantation , Neoplasms/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Skin Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...