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.
Am J Physiol Endocrinol Metab ; 292(3): E964-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17132826

ABSTRACT

We generated three fully human monoclonal antibody antagonists against fibroblast growth factor receptor-1 (FGFR1) that potently block FGF signaling. We found that antibodies targeting the c-splice form of the receptor (FGFR1c) were anorexigenic when administered intraperitoneally three times weekly to mice, resulting in rapid, dose-dependent weight loss that plateaued (for doses>4 mg/kg) at 35-40% in 2 wk. Animals appeared healthy during treatment and regained their normal body weights and growth trajectories upon clearance of the antibodies from the bloodstream. Measurements of food consumption and energy expenditure indicated that the rapid weight loss was induced primarily by decreased energy intake and not by increased energy expenditure or cachexia and was accompanied by a greater reduction in fat than lean body mass. Hypophagia was not caused through malaise or illness, as indicated by absence of conditioned taste aversion, pica behavior, and decreased need-induced salt intake in rats. In support of a hypothalamic site of action, we found that, after intraperitoneal injections, anti-FGFR1c (IMC-A1), but not a control antibody, accumulated in the median eminence and adjacent mediobasal hypothalamus and that FGFR1c is enriched in the hypothalamus of mice. Furthermore, a single intracerebroventricular administration of 3 microg of IMC-A1 via the 3rd ventricle to mice caused an approximately 36% reduction in food intake and an approximately 6% weight loss within the ensuing 24 h. Our data suggest that FGF signaling through FGFR1c may play a physiological role in hypothalamic feeding circuit and that blocking it leads to hypophagia and weight loss.


Subject(s)
Antibodies, Monoclonal/pharmacology , Eating/drug effects , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/immunology , Weight Loss/drug effects , Animals , Antibodies, Monoclonal/adverse effects , Antibody Specificity , Body Composition/drug effects , Central Nervous System/drug effects , Energy Metabolism/drug effects , Female , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/immunology , Rats , Rats, Sprague-Dawley
2.
Clin Cancer Res ; 12(7 Pt 1): 2197-207, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16609035

ABSTRACT

PURPOSE: Combination therapies that target the epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor (VEGFR) pathways, are being actively tested for the treatment of cancer. In evaluating combination strategies, the ideal combination would be one in which the treatments interact in a way that is synergistic with regard to antitumor effects. Here, we have evaluated the interaction between anti-EGFR antibody Erbitux (cetuximab) and anti-VEGFR2 antibody, DC101, in preclinical models of pancreatic (BxPC-3) and colon (GEO) cancer. EXPERIMENTAL DESIGN: Analysis of the interaction between cetuximab and DC101 in vivo used a novel method for establishing the upper 95% confidence limits for the combination index (CI) of isobologram analyses, where CI < 1 indicates synergy. Assessment of tumor cell proliferation, apoptosis, VEGF production, and hypoxia, as well as tumor vascularization, was performed to gain insights into the mechanistic basis for synergy between agents targeting different tumor compartments. RESULTS: Monotherapy ED(50) values for tumor growth inhibition ranged from 1.8 to 2.3 mg/kg and 10.5 to 16.6 mg/kg for cetuximab and DC101, respectively. From the dose response of the combination treatment, it was determined that cetuximab and DC101 are synergistic in the BxPC-3 (CI = 0.1, P < 0.01) and GEO (CI = 0.1, P < 0.01) models. Overlapping effects on the tumor cell and vascular compartments form a basis for the interaction, with VEGF production and hypoxia-inducible factor 1alpha potentially acting as molecular links between EGFR and VEGFR2 inhibition. CONCLUSIONS: Results show antitumor synergy for combined EGFR and VEGFR2 targeted therapy, supporting the significant therapeutic potential of this combination strategy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Colonic Neoplasms/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Structure-Activity Relationship , Transplantation, Heterologous , Xenograft Model Antitumor Assays
3.
Circulation ; 110(16): 2430-5, 2004 Oct 19.
Article in English | MEDLINE | ID: mdl-15477421

ABSTRACT

BACKGROUND: The rate of reendothelialization is critical in neointima formation after arterial injury. Vascular endothelial growth factor (VEGF), a potent endothelial mitogen, has been advocated for accelerating endothelial repair and preventing intimal hyperplasia after percutaneous coronary interventions. However, the precise mechanism of action of VEGF treatment and the physiologic role of endogenous VEGF after arterial injury are not well described. To better understand the role of VEGF in arterial repair, we overexpressed both VEGF and a soluble, chimeric VEGF receptor (VEGF-trap), which binds free VEGF with high affinity, in a mouse model of arterial injury. METHODS AND RESULTS: Four groups of C57BL/6 mice underwent denuding endothelial injury 1 day after systemic injection of recombinant adenovirus expressing (1) VEGF, (2) VEGF-trap, (3) VEGF plus VEGF-trap, or (4) control adenovirus. Circulating levels of adenovirus-encoded proteins were significantly elevated after gene transfer. VEGF overexpression accelerated reendothelialization and increased luminal endothelial cell proliferation 2 weeks after arterial injury (P<0.05), resulting in decreased neointima formation at 4 weeks compared with control (P<0.01). Cotreatment with VEGF-trap completely sequestered free VEGF and abrogated the beneficial effect of VEGF overexpression. Interestingly, sequestration of endogenous VEGF by VEGF-trap overexpression alone also led to delayed reendothelialization at 2 weeks (P<0.01) and increased neointima formation at 4 weeks (P<0.01). CONCLUSIONS: VEGF overexpression accelerated endothelial repair and inhibited neointima formation after arterial injury. Conversely, sequestration of exogenous and/or endogenous VEGF by VEGF-trap delayed reendothelialization and significantly increased neointima size. This demonstrates the therapeutic potential of VEGF but also emphasizes the important physiologic role of endogenous VEGF in vascular repair.


Subject(s)
Endothelium, Vascular/injuries , Genetic Therapy , Vascular Endothelial Growth Factor A/physiology , Wound Healing/physiology , Angioplasty/adverse effects , Animals , Cell Division , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Humans , Hyperplasia , Image Processing, Computer-Assisted , Male , Mice , Mice, Inbred C57BL , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Single-Blind Method , Tunica Intima/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
4.
Circulation ; 109(16): 2001-8, 2004 Apr 27.
Article in English | MEDLINE | ID: mdl-15078795

ABSTRACT

BACKGROUND: Macrophages associated with arterial wall lipid deposition contribute to inflammatory processes. Tissue factor (TF) has been implicated in the thrombogenicity of atherosclerotic plaques. Intimal cells undergoing apoptosis have been postulated as a source for TF. However, there is only limited knowledge of cell type, plaque component, and conditions associated with TF expression and apoptosis. We examined the hypothesis that macrophages exposed to conditions of lipid-rich plaque undergo apoptosis and express TF. METHODS AND RESULTS: In human carotid (n=15) and coronary (n=6) atherosclerotic plaques, TF and caspase-3 mRNA and protein expression (evaluated by in situ hybridization and immunohistochemistry) were increased significantly in lipid-rich compared with fibrous plaque components (P<0.01) and correlated with high macrophage content (P<0.05). Double-labeling studies demonstrated colocalization of TF and active caspase-3. In hyperlipidemic mice, expression of TF and active caspase-3 was observed simultaneously and colocalized in neointimal macrophages after arterial injury. In neointima of normolipidemic animals, TF and active caspase-3 were absent after arterial injury. In monocytes cultured in the presence of oxidized LDL, strong induction and colocalization of TF and active caspase-3 were found compared with baseline (P<0.05). Both antigens were significantly decreased after cotreatment with a caspase inhibitor (P<0.05) and were absent in untreated control cells. CONCLUSIONS: The expression of TF as the primary cell-associated activator of the coagulation pathway proves to be closely related to macrophages undergoing apoptosis in conditions of lipid-rich plaque, pointing to a key role of lipid content and inflammatory cell viability in determining plaque thrombogenicity.


Subject(s)
Apoptosis , Arteriosclerosis/metabolism , Caspases/metabolism , Foam Cells/metabolism , Thromboplastin/metabolism , Animals , Arteriosclerosis/immunology , Arteriosclerosis/pathology , Caspase 3 , Caspases/genetics , Foam Cells/enzymology , Foam Cells/pathology , Gene Expression , Humans , Inflammation/complications , Male , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Thromboplastin/genetics , Thrombosis/immunology
5.
J Hepatol ; 40(2): 305-12, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14739103

ABSTRACT

BACKGROUND/AIMS: Liver regeneration is dependent upon coordinated proliferation of hepatocytes and endothelial cells. Vascular endothelial growth factor (VEGF) promotes angiogenesis. Hepatic steatosis delays regeneration and increases liver resection morbidity. We hypothesized that VEGF overexpression stimulates hepatic regeneration. METHODS: Recombinant adenovirus expressing human VEGF165 or adenovirus control-vector (LacZ) were administered before 2/3 hepatectomy in lean and ob/ob mice. Galactose elimination capacity, a quantitative liver function test, was repeatedly measured before and after hepatectomy. Expression of VEGF receptors (flt1, flk1), endoglin and hypoxia inducible factor-1alpha (HIF-1alpha) was assessed by quantitative RT-PCR and for endoglin also by immunohistochemistry. RESULTS: After 2/3 hepatectomy, VEGF gene transfer increased galactose elimination capacity in lean and ob/ob mice. HIF-1alpha, endoglin and VEGF receptor mRNA increased during regeneration in lean but not in obese mice. Staining of endothelial cells by endoglin immunohistochemistry returned to baseline reactivity in lean mice by day 6 and remained decreased in ob/ob mice. VEGF treatment decreased HIF-1alpha and increased flk1 response in lean mice. CONCLUSIONS: Hepatic resection elicits an angiogenic response in the remnant liver, which is impaired in case of steatosis. Adenovirus-mediated transfer of VEGF hastens functional hepatic recovery in lean, and more importantly also, in obese mice after partial hepatectomy.


Subject(s)
Fatty Liver/physiopathology , Fatty Liver/therapy , Genetic Therapy , Hepatectomy , Obesity/physiopathology , Vascular Endothelial Growth Factor A/genetics , Adenoviridae/genetics , Animals , Antigens, CD , Body Weight , Breath Tests , Endoglin , Fatty Liver/pathology , Galactose/metabolism , Hepatocytes/physiology , Hypoxia-Inducible Factor 1, alpha Subunit , Lac Operon , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Organ Size , RNA, Messenger/analysis , Receptors, Cell Surface , Recovery of Function , Transcription Factors/genetics , Transgenes , Vascular Cell Adhesion Molecule-1/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Circulation ; 107(12): 1658-63, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12668502

ABSTRACT

BACKGROUND: Impaired endothelial regeneration contributes to arterial lesion formation. Endostatin is a specific inhibitor of endothelial cell growth and induces endothelial cell apoptosis. We examined the effect of endostatin overexpression on reendothelialization and neointima formation in a mouse model of arterial injury. METHODS AND RESULTS: Mice underwent femoral arterial denudation and received recombinant adenovirus, expressing either murine endostatin (n=19) or control adenoviral vector (n=12), by jugular vein injection. Endostatin gene transfer resulted in high serum levels of endostatin. Strong adenoviral gene expression of beta-galactosidase-expressing control vector was detected in liver tissue and was absent in the injured arterial wall at 1 week. Deposits of endostatin protein were detected along the denuded arterial wall and were not seen in the noninjured contralateral artery at 1 week. Endostatin deposits were also absent in the injured artery of control vector-treated animals. Overexpression of endostatin led to decreased reendothelialization and increased apoptosis of luminal endothelial cells 2 and 4 weeks after arterial injury (P<0.05). In addition, endostatin overexpression resulted in increased neointima formation (P<0.05). Endothelial apoptosis and neointima area correlated positively with endostatin serum levels, whereas the degree of reendothelialization correlated negatively with endostatin serum levels (P<0.05). Furthermore, poor reendothelialization correlated with increased neointima formation (P<0.05). CONCLUSIONS: In summary, decreased reendothelialization and enhanced endothelial apoptosis, in response to endostatin overexpression, were associated with increased neointima formation. These findings demonstrate that high serum levels of endostatin are capable of inhibiting endothelial regeneration and promoting arterial lesion growth in conditions of endothelial injury.


Subject(s)
Arterial Occlusive Diseases/etiology , Collagen/physiology , Endothelium, Vascular/physiology , Peptide Fragments/physiology , Adenoviridae/genetics , Animals , Apoptosis , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Collagen/genetics , Collagen/metabolism , Endostatins , Endothelium, Vascular/pathology , Gene Expression , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/genetics , Peptide Fragments/metabolism , Transgenes , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL