Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neoplasia ; 15(7): 694-711, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23814482

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway is a central mediator of vascular endothelial growth factor (VEGF)-driven angiogenesis. The discovery of small molecule inhibitors that selectively target PI3K or PI3K and mammalian target of rapamycin (mTOR) provides an opportunity to pharmacologically determine the contribution of these key signaling nodes in VEGF-A-driven tumor angiogenesis in vivo. This study used an array of micro-vascular imaging techniques to monitor the antivascular effects of selective class I PI3K, mTOR, or dual PI3K/mTOR inhibitors in colorectal and prostate cancer xenograft models. Micro-computed tomography (micro-CT) angiography, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), vessel size index (VSI) MRI, and DCE ultrasound (DCE-U/S) were employed to quantitatively evaluate the vascular (structural and physiological) response to these inhibitors. GDC-0980, a dual PI3K/mTOR inhibitor, was found to reduce micro-CT angiography vascular density, while VSI MRI demonstrated a significant reduction in vessel density and an increase in mean vessel size, consistent with a loss of small functional vessels and a substantial antivascular response. DCE-MRI showed that GDC-0980 produces a strong functional response by decreasing the vascular permeability/perfusion-related parameter, K (trans). Interestingly, comparable antivascular effects were observed for both GDC-980 and GNE-490 (a selective class I PI3K inhibitor). In addition, mTOR-selective inhibitors did not affect vascular density, suggesting that PI3K inhibition is sufficient to generate structural changes, characteristic of a robust antivascular response. This study supports the use of noninvasive microvascular imaging techniques (DCE-MRI, VSI MRI, DCE-U/S) as pharmacodynamic assays to quantitatively measure the activity of PI3K and dual PI3K/mTOR inhibitors in vivo.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors , Neoplasms/diagnosis , Neovascularization, Pathologic/diagnosis , Angiography/methods , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterografts , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Magnetic Resonance Imaging/methods , Mice , Multimodal Imaging , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Burden/drug effects , Ultrasonography/methods , X-Ray Microtomography/methods
2.
PLoS One ; 8(7): e68755, 2013.
Article in English | MEDLINE | ID: mdl-23874750

ABSTRACT

Tumor-associated lymphatics are postulated to provide a transit route for disseminating metastatic cells. This notion is supported by preclinical findings that inhibition of pro-lymphangiogenic signaling during tumor development reduces cell spread to sentinel lymph nodes (SLNs). However, it is unclear how lymphatics downstream of SLNs contribute to metastatic spread into distal organs, or if modulating distal lymph transport impacts disease progression. Utilizing murine models of metastasis, longitudinal in vivo imaging of lymph transport, and function blocking antibodies against two VEGF family members, we provide evidence that distal lymphatics undergo disease course-dependent up-regulation of lymph transport coincidental with structural remodeling. Inhibition of VEGF-C activity with antibodies against VEGF-C or NRP2 prevented these disease-associated changes. Furthermore, utilizing a novel model of adjuvant treatment, we demonstrate that antagonism of VEGF-C or NRP2 decreases post SLN metastasis. These data support a potential therapeutic strategy for inhibiting distant metastatic dissemination via targeting tumor-associated lymphatic remodeling.


Subject(s)
Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Vascular Endothelial Growth Factor C/metabolism , Animals , Antibodies/therapeutic use , Female , Lymph/physiology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphatic Metastasis/physiopathology , Lymphatic Metastasis/prevention & control , Male , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Vascular Endothelial Growth Factor C/antagonists & inhibitors
3.
Circ Res ; 109(5): 486-91, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21778431

ABSTRACT

RATIONALE: Collagen- and calcium-binding EGF domains 1 (CCBE1) has been associated with Hennekam syndrome, in which patients have lymphedema, lymphangiectasias, and other cardiovascular anomalies. Insight into the molecular role of CCBE1 is completely lacking, and mouse models for the disease do not exist. OBJECTIVE: CCBE1 deficient mice were generated to understand the function of CCBE1 in cardiovascular development, and CCBE1 recombinant protein was used in both in vivo and in vitro settings to gain insight into the molecular function of CCBE1. METHODS AND RESULTS: Phenotypic analysis of murine Ccbe1 mutant embryos showed a complete lack of definitive lymphatic structures, even though Prox1(+) lymphatic endothelial cells get specified within the cardinal vein. Mutant mice die prenatally. Proximity ligation assays indicate that vascular endothelial growth factor receptor 3 activation appears unaltered in mutants. Human CCBE1 protein binds to components of the extracellular matrix in vitro, and CCBE1 protein strongly enhances vascular endothelial growth factor-C-mediated lymphangiogenesis in a corneal micropocket assay. CONCLUSIONS: Our data identify CCBE1 as a factor critically required for budding and migration of Prox-1(+) lymphatic endothelial cells from the cardinal vein. CCBE1 probably exerts these effects through binding to components of the extracellular matrix. CCBE1 has little lymphangiogenic effect on its own but dramatically enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo. Thus, our data suggest CCBE1 to be essential but not sufficient for lymphangiogenesis.


Subject(s)
Calcium-Binding Proteins/physiology , Endothelium, Lymphatic/blood supply , Endothelium, Lymphatic/metabolism , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Lymphatic Vessels/metabolism , Tumor Suppressor Proteins/physiology , Vascular Endothelial Growth Factor C/metabolism , Animals , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cornea/blood supply , Cornea/cytology , Cornea/metabolism , Endothelium, Lymphatic/cytology , Humans , Lymphangiogenesis/genetics , Mice , Mice, Knockout , Protein Binding/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/physiology
4.
J Cell Biol ; 188(1): 115-30, 2010 Jan 11.
Article in English | MEDLINE | ID: mdl-20065093

ABSTRACT

Vascular sprouting is a key process-driving development of the vascular system. In this study, we show that neuropilin-2 (Nrp2), a transmembrane receptor for the lymphangiogenic vascular endothelial growth factor C (VEGF-C), plays an important role in lymphatic vessel sprouting. Blocking VEGF-C binding to Nrp2 using antibodies specifically inhibits sprouting of developing lymphatic endothelial tip cells in vivo. In vitro analyses show that Nrp2 modulates lymphatic endothelial tip cell extension and prevents tip cell stalling and retraction during vascular sprout formation. Genetic deletion of Nrp2 reproduces the sprouting defects seen after antibody treatment. To investigate whether this defect depends on Nrp2 interaction with VEGF receptor 2 (VEGFR2) and/or 3, we intercrossed heterozygous mice lacking one allele of these receptors. Double-heterozygous nrp2vegfr2 mice develop normally without detectable lymphatic sprouting defects. In contrast, double-heterozygote nrp2vegfr3 mice show a reduction of lymphatic vessel sprouting and decreased lymph vessel branching in adult organs. Thus, interaction between Nrp2 and VEGFR3 mediates proper lymphatic vessel sprouting in response to VEGF-C.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Neuropilin-2/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cell Shape , Cells, Cultured , Female , Lymphangiogenesis , Lymphatic Vessels/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Neuropilin-2/genetics , Protein Binding , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics
5.
Cancer Cell ; 13(4): 331-42, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18394556

ABSTRACT

Metastasis, which commonly uses lymphatics, accounts for much of the mortality associated with cancer. The vascular endothelial growth factor (VEGF)-C coreceptor, neuropilin-2 (Nrp2), modulates but is not necessary for developmental lymphangiogenesis, and its significance for metastasis is unknown. An antibody to Nrp2 that blocks VEGFC binding disrupts VEGFC-induced lymphatic endothelial cell migration, but not proliferation, in part independently of VEGF receptor activation. It does not affect established lymphatics in normal adult mice but reduces tumoral lymphangiogenesis and, importantly, functional lymphatics associated with tumors. It also reduces metastasis to sentinel lymph nodes and distant organs, apparently by delaying the departure of tumor cells from the primary tumor. Our results demonstrate that Nrp2, which was originally identified as an axon-guidance receptor, is an attractive target for modulating metastasis.


Subject(s)
Neoplasm Metastasis/prevention & control , Neoplasms/pathology , Neuropilin-2/antagonists & inhibitors , Animals , Antibodies, Blocking/pharmacology , Antibody Specificity/drug effects , Bacteriophages , Cell Line , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphangiogenesis/drug effects , Lymphatic Metastasis/prevention & control , Lymphatic System/drug effects , Lymphatic System/pathology , Mice , Neuropilin-2/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xenograft Model Antitumor Assays
6.
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
7.
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
8.
Int J Radiat Oncol Biol Phys ; 65(2): 517-27, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16690434

ABSTRACT

PURPOSE: To determine whether para-aminobenzoic acid (PABA) alters the sensitivity of tumor cells to ionizing radiation in vitro and in vivo. METHODS AND MATERIALS: Cellular proliferation was assessed by WST-1 assays. The effects of PABA and radiation on tumor growth were examined with chick embryo and murine models. Real-time reverse transcriptase-polymerase chain reaction and Western blotting were used to quantify p21CIP1 and CDC25A levels. RESULTS: Para-aminobenzoic acid enhanced (by 50%) the growth inhibitory activity of radiation on B16F10 cells, whereas it had no effect on melanocytes. Para-aminobenzoic acid enhanced (50-80%) the antitumor activity of radiation on B16F10 and 4T1 tumors in vivo. The combination of PABA and radiation therapy increased tumor apoptosis. Treatment of tumor cells with PABA increased expression of CDC25A and decreased levels of p21CIP1. CONCLUSIONS: Our findings suggest that PABA might represent a compound capable of enhancing the antitumor activity of ionizing radiation by a mechanism involving altered expression of proteins known to regulate cell cycle arrest.


Subject(s)
4-Aminobenzoic Acid/therapeutic use , Dietary Supplements , Melanocytes/drug effects , Melanocytes/radiation effects , Melanoma/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Animals , Apoptosis/radiation effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Chick Embryo , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymerase Chain Reaction
9.
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
10.
Cancer Res ; 66(8): 4125-32, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16618733

ABSTRACT

The mechanism of thrombin-induced angiogenesis is poorly understood. Using a gene chip array to investigate the pro-malignant phenotype of thrombin-stimulated cells, we observed that thrombin markedly up-regulates growth-regulated oncogene-alpha (GRO-alpha) in several tumor cell lines as well as endothelial cells by mRNA and protein analysis. Thrombin enhanced the secretion of GRO-alpha from tumor cells 25- to 64-fold. GRO-alpha is a CXC chemokine with tumor-associated angiogenic as well as oncogenic activation following ligation of its CXCR2 receptor. GRO-alpha enhanced angiogenesis in the chick chorioallantoic membrane assay 2.2-fold, providing direct evidence for GRO-alpha as an angiogenic growth factor. Anti-GRO-alpha antibody completely inhibited the 2.7-fold thrombin-induced up-regulation of angiogenesis, as well as the 1.5-fold thrombin-induced up-regulation of both endothelial cell cord formation in Matrigel and growth in vitro. Thrombin as well as its PAR-1 receptor activation peptide [thrombin receptor activation peptide (TRAP)] as well as GRO-alpha all markedly increased vascular regulatory proteins and growth factors: matrix metalloproteinase (MMP)-1, MMP-2, vascular endothelial growth factor (VEGF), angiopoietin-2 (Ang-2), CD31, and receptors KDR and CXCR2 in human umbilical vein endothelial cells. All of the thrombin/TRAP gene up-regulations were completely inhibited by anti-GRO-alpha antibody and unaffected by irrelevant antibody. Similar inhibition of gene up-regulation as well as thrombin-induced chemotaxis was noted with small interfering RNA (shRNA) GRO-alpha KD 4T1 breast tumor and B16F10 melanoma cells. In vivo tumor growth studies in wild-type mice with shRNA GRO-alpha KD cells revealed 2- to 4-fold impaired tumor growth, metastasis, and angiogenesis, which was not affected by endogenous thrombin. Thus, thrombin-induced angiogenesis requires the up-regulation of GRO-alpha. Thrombin up-regulation of GRO-alpha in tumor cells as well as endothelial cells contributes to tumor angiogenesis.


Subject(s)
Chemokines, CXC/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Neoplasms/blood supply , Neovascularization, Physiologic/physiology , Thrombin/pharmacology , Amino Acid Sequence , Angiopoietin-2/biosynthesis , Animals , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Chemokine CXCL1 , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Chick Embryo , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Matrix Metalloproteinase 1/biosynthesis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Thrombin/antagonists & inhibitors , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...