Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006633

ABSTRACT

Neovascularization is a key feature of ischemic retinal diseases and the wet form of age-related macular degeneration (AMD), all leading causes of severe vision loss. Vascular endothelial growth factor (VEGF) inhibitors have transformed the treatment of these disorders. Millions of patients have been treated with these drugs worldwide. However, in real-life clinical settings, many patients do not experience the same degree of benefit observed in clinical trials, in part because they receive fewer anti-VEGF injections. Therefore, there is an urgent need to discover and identify novel long-acting VEGF inhibitors. We hypothesized that binding to heparan-sulfate proteoglycans (HSPG) in the vitreous, and possibly other ocular structures, may be a strategy to promote intraocular retention, ultimately leading to a reduced burden of intravitreal injections. We designed a series of VEGF receptor 1 variants and identified some with strong heparin-binding characteristics and ability to bind to vitreous matrix. Our data indicate that some of our variants have longer duration and greater efficacy in animal models of intraocular neovascularization than current standard of care. Our study represents a systematic attempt to exploit the functional diversity associated with heparin affinity of a VEGF receptor.


Subject(s)
Choroidal Neovascularization/drug therapy , Heparan Sulfate Proteoglycans/pharmacology , Macular Degeneration/drug therapy , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Angiogenesis Inhibitors/chemistry , Animals , Cell Proliferation/drug effects , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Crystallography, X-Ray , Endothelial Cells/drug effects , Eye/drug effects , Eye/pathology , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/immunology , Heparin/genetics , Human Umbilical Vein Endothelial Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/ultrastructure , Intravitreal Injections , Macular Degeneration/genetics , Macular Degeneration/pathology , Mice , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/ultrastructure , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vitreous Body/drug effects
2.
Nat Commun ; 11(1): 6330, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303737

ABSTRACT

Endothelial cell (EC) metabolism is thought to be one of the driving forces for angiogenesis. Here we report the identification of the hexosamine D-mannosamine (ManN) as an EC mitogen and survival factor for bovine and human microvascular EC, with an additivity with VEGF. ManN inhibits glycosylation in ECs and induces significant changes in N-glycan and O-glycan profiles. We further demonstrate that ManN and two N-glycosylation inhibitors stimulate EC proliferation via both JNK activation and the unfolded protein response caused by ER stress. ManN results in enhanced angiogenesis in a mouse skin injury model. ManN also promotes angiogenesis in a mouse hindlimb ischemia model, with accelerated limb blood flow recovery compared to controls. In addition, intraocular injection of ManN induces retinal neovascularization. Therefore, activation of stress pathways following inhibition of protein glycosylation can promote EC proliferation and angiogenesis and may represent a therapeutic strategy for treatment of ischemic disorders.


Subject(s)
Neovascularization, Physiologic , Proteins/metabolism , Stress, Physiological , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Female , Glycosylation/drug effects , Heat-Shock Proteins/metabolism , Hexosamines/pharmacology , Hindlimb/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/pathology , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Microvessels/metabolism , Neovascularization, Physiologic/drug effects , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Skin/pathology , Stress, Physiological/drug effects , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing/drug effects
3.
Proc Natl Acad Sci U S A ; 117(35): 21598-21608, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817421

ABSTRACT

We tested cis-ApcΔ716/Smad4+/- and cis-ApcΔ716/Smad4+/-KrasG12D mice, which recapitulate key genetic abnormalities accumulating during colorectal cancer (CRC) tumorigenesis in humans, for responsiveness to anti-VEGF therapy. We found that even tumors in cis-ApcΔ716/Smad4+/-KrasG12D mice, although highly aggressive, were suppressed by anti-VEGF treatment. We tested the hypothesis that inflammation, a major risk factor and trigger for CRC, may affect responsiveness to anti-VEGF. Chemically induced colitis (CIC) in cis-ApcΔ716/Smad4+/- and cis-ApcΔ716/Smad4+/-KrasG12D mice promoted development of colon tumors that were largely resistant to anti-VEGF treatment. The myeloid growth factor G-CSF was markedly increased in the serum after induction of colitis. Antibodies blocking G-CSF, or its target Bv8/PROK2, suppressed tumor progression and myeloid cell infiltration when combined with anti-VEGF in CIC-associated CRC and in anti-VEGF-resistant CRC liver metastasis models. In a series of CRC specimens, tumor-infiltrating neutrophils strongly expressed Bv8/PROK2. CRC patients had significantly higher plasma Bv8/PROK2 levels than healthy volunteers and high plasma Bv8/PROK2 levels were inversely correlated with overall survival. Our findings establish Bv8/PROK2 as a translational target in CRC, in combination with anti-VEGF agents.


Subject(s)
Colorectal Neoplasms/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Antibodies/immunology , Colonic Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Female , Granulocyte Colony-Stimulating Factor/metabolism , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Myeloid Cells/metabolism , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
4.
Cell ; 167(1): 275-284.e6, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662093

ABSTRACT

The VEGF-A isoforms play a crucial role in vascular development, and the VEGF signaling pathway is a clinically validated therapeutic target for several pathological conditions. Alternative mRNA splicing leads to the generation of multiple VEGF-A isoforms, including VEGF165. A recent study reported the presence of another isoform, VEGF-Ax, arising from programmed readthrough translation. Compared to VEGF165, VEGF-Ax has a 22-amino-acid extension in the COOH terminus and has been reported to function as a negative regulator of VEGF signaling in endothelial cells, with potent anti-angiogenic effects. Here, we show that, contrary to the earlier report, VEGF-Ax stimulates endothelial cell mitogenesis, angiogenesis, as well as vascular permeability. Accordingly, VEGF-Ax induces phosphorylation of key tyrosine residues in VEGFR-2. Notably, VEGF-Ax was less potent than VEGF165, consistent with its impaired binding to the VEGF co-receptor neuropilin-1.


Subject(s)
Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A , Alternative Splicing , Amino Acid Sequence , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Capillary Permeability/genetics , Capillary Permeability/physiology , Chemotaxis/drug effects , Cloning, Molecular , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Guinea Pigs , HEK293 Cells , Humans , Mice , Mitogens/pharmacology , Mitosis/drug effects , Mitosis/physiology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Neuropilin-1/metabolism , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Clin Cancer Res ; 19(16): 4433-45, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23812669

ABSTRACT

PURPOSE: Our goal was to develop a potent humanized antibody against mouse/human CXCL12. This report summarized its in vitro and in vivo activities. EXPERIMENTAL DESIGN: Cell surface binding and cell migration assays were used to select neutralizing hamster antibodies, followed by testing in several animal models. Monoclonal antibody (mAb) 30D8 was selected for humanization based on its in vitro and in vivo activities. RESULTS: 30D8, a hamster antibody against mouse and human CXCL12α, CXCL12ß, and CXCL12γ, was shown to dose-dependently block CXCL12α binding to CXCR4 and CXCR7, and CXCL12α-induced Jurkat cell migration in vitro. Inhibition of primary tumor growth and/or metastasis was observed in several models. 30D8 alone significantly ameliorated arthritis in a mouse collagen-induced arthritis model (CIA). Combination with a TNF-α antagonist was additive. In addition, 30D8 inhibited 50% of laser-induced choroidal neovascularization (CNV) in mice. Humanized 30D8 (hu30D8) showed similar in vitro and in vivo activities as the parental hamster antibody. A crystal structure of the hu30D8 Fab/CXCL12α complex in combination with mutational analysis revealed a "hot spot" around residues Asn(44)/Asn(45) of CXCL12α and part of the RFFESH region required for CXCL12α binding to CXCR4 and CXCR7. Finally, hu30D8 exhibited fast clearance in cynomolgus monkeys but not in rats. CONCLUSION: CXCL12 is an attractive target for treatment of cancer and inflammation-related diseases; hu30D8 is suitable for testing this hypothesis in humans.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Chemokine CXCL12/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Cell Line, Tumor , Chemokine CXCL12/chemistry , Chemokine CXCL12/metabolism , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Epitope Mapping , Female , Humans , Mice , Models, Molecular , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Protein Conformation , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
6.
PLoS One ; 5(1): e8611, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20087418

ABSTRACT

BACKGROUND: Histological examinations of MMTV-Wnt1 tumors reveal drastic differences in the tumor vasculature when compared to MMTV-Her2 tumors. However, these differences have not been formally described, nor have any angiogenic factors been implicated to be involved in the Wnt1 tumors. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that MMTV-Wnt1 tumors were more vascularized than MMTV-Her2 tumors, and this correlated with significantly higher expression of a CXC chemokine, stromal cell-derived factor-1 (SDF1/CXCL12) but not with VEGFA. Isolation of various cell types from Wnt1 tumors revealed that SDF1 was produced by both tumor myoepithelial cells and stromal cells, whereas Her2 tumors lacked myoepithelial cells and contained significantly less stroma. The growth of Wnt1 tumors, but not Her2 tumors, was inhibited by a neutralizing antibody to SDF1, but not by neutralization of VEGFA. Anti-SDF1 treatment decreased the proportion of infiltrating Gr1(+) myeloid cells in the Wnt1 tumors, which correlated with a decrease in the percentage of endothelial cells. The involvement of Gr1(+) cells was evident from the retardation of Wnt1 tumor growth following in vivo depletion of these cells with an anti-Gr1-specific antibody. This degree of inhibition on Wnt1 tumor growth was comparable, but not additive, to the effect observed with anti-SDF1, indicative of overlapping mechanisms of inhibition. In contrast, Her2 tumors were not affected by the depletion of Gr1(+) cells. CONCLUSIONS/SIGNIFICANCE: We demonstrated that SDF1 is important for Wnt1, but not for HER2, in inducing murine mammary tumor and the role of SDF1 in tumorigenesis involves Gr1(+) myeloid cells to facilitate growth and/or angiogenesis.


Subject(s)
CD11b Antigen/physiology , Chemokine CXCL12/physiology , Mammary Tumor Virus, Mouse/physiology , Neoplasms, Experimental/physiopathology , Wnt1 Protein/physiology , Animals , Mice , Neoplasms, Experimental/virology , Vascular Endothelial Growth Factor A/physiology
7.
Clin Cancer Res ; 15(8): 2675-84, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19336519

ABSTRACT

PURPOSE: Bv8, also known as prokineticin 2, has been recently shown to be a mediator of myeloid cell-dependent tumor angiogenesis in mouse models. We wished to determine whether these findings might be potentially relevant to human disease. EXPERIMENTAL DESIGN: We characterized Bv8 expression in human blood cells in vitro and in vivo, and did Bv8 immunohistochemistry in human tumor sections. We also partially purified Bv8 from human neutrophils and tested its bioactivity. RESULTS: We found that Bv8 expression is regulated by several cytokines in a cell type-specific fashion. Both granulocyte colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor induced Bv8 expression in neutrophils and bone marrow cells, whereas interleukin 10 up-regulated Bv8 expression in monocytes and lymphocytes. Bv8 potently promoted neutrophil chemotaxis. Bv8 protein isolated from human neutrophils was found to be biologically active. Of the two receptors for Bv8 [prokineticin receptor 1(PKR1)/endocrine gland-derived vascular endothelial growth factor receptor 1 (EG-VEGFR1) and PKR2/EG-VEGFR2], only PKR2/EG-VEGFR2 was detectable in human neutrophils. Also, we found a marked up-regulation of Bv8 mRNA and protein in peripheral blood mononuclear cells from G-CSF-treated donors compared with those from untreated individuals, verifying our in vitro observations. Finally, immunohistochemistry showed Bv8 expression in neutrophils infiltrating human tumors. CONCLUSIONS: These results provide the basis for further investigation of the pathophysiologic role of Bv8 in human tumors and inflammatory disorders and, potentially, for therapeutic application of Bv8 inhibitors.


Subject(s)
Gastrointestinal Hormones/blood , Monocytes/metabolism , Neuropeptides/blood , Neutrophils/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/pharmacology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Neutrophils/drug effects , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Cancer Cell ; 15(1): 21-34, 2009 Jan 06.
Article in English | MEDLINE | ID: mdl-19111878

ABSTRACT

Tumor- or cancer-associated fibroblasts (TAFs or CAFs) from different tumors exhibit distinct angiogenic and tumorigenic properties. Unlike normal skin fibroblasts or TAFs from TIB6 tumors that are sensitive to anti-VEGF treatment (TAF-TIB6), TAFs from resistant EL4 tumors (TAF-EL4) can stimulate TIB6 tumor growth even when VEGF is inhibited. We show that platelet-derived growth factor C (PDGF-C) is upregulated in TAFs from resistant tumors. PDGF-C-neutralizing antibodies blocked the angiogenesis induced by such TAFs in vivo, slowed the growth of EL4 and admixture (TAF-EL4 + TIB6) tumors, and exhibited additive effects with anti-VEGF-A antibodies. Hence, our data reveal an additional mechanism for TAF-mediated tumorigenesis and suggest that some tumors may overcome inhibition of VEGF-mediated angiogenesis through upregulation of PDGF-C.


Subject(s)
Antibodies/therapeutic use , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Fibroblasts/metabolism , Lymphokines/metabolism , Neovascularization, Pathologic/metabolism , Platelet-Derived Growth Factor/metabolism , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies/immunology , CD11b Antigen/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Separation , Cell Transformation, Neoplastic/immunology , Cells, Cultured , Chemokine CXCL12/metabolism , Disease Progression , Drug Resistance, Neoplasm/drug effects , Fibroblasts/pathology , Humans , Immunotherapy , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Up-Regulation
9.
Trends Cell Biol ; 18(8): 372-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18614368

ABSTRACT

Cells of the innate immune system have a key role in maintaining homeostasis by providing the first line of defense against many pathogens. Innate immunity can also modulate the activity of acquired immunity by several mechanisms. However, subsets of myeloid cells can facilitate tumor growth, because these cells produce angiogenic factors and can also prevent the immune system from attacking tumor cells. Recent studies also emphasize the role of myeloid cells in mediating refractoriness to anti-VEGF treatments. This function of myeloid cells occurs through a proangiogenic pathway that is, at least in part, driven by the secreted protein Bv8. This review summarizes recent findings on the complex role of bone marrow-derived cells in tumor growth.


Subject(s)
Myeloid Cells/pathology , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Bone Marrow Cells/pathology , Humans , Tumor Escape
10.
Nature ; 450(7171): 825-31, 2007 Dec 06.
Article in English | MEDLINE | ID: mdl-18064003

ABSTRACT

Bone-marrow-derived cells facilitate tumour angiogenesis, but the molecular mechanisms of this facilitation are incompletely understood. We have previously shown that the related EG-VEGF and Bv8 proteins, also known as prokineticin 1 (Prok1) and prokineticin 2 (Prok2), promote both tissue-specific angiogenesis and haematopoietic cell mobilization. Unlike EG-VEGF, Bv8 is expressed in the bone marrow. Here we show that implantation of tumour cells in mice resulted in upregulation of Bv8 in CD11b+Gr1+ myeloid cells. We identified granulocyte colony-stimulating factor as a major positive regulator of Bv8 expression. Anti-Bv8 antibodies reduced CD11b+Gr1+ cell mobilization elicited by granulocyte colony-stimulating factor. Adenoviral delivery of Bv8 into tumours was shown to promote angiogenesis. Anti-Bv8 antibodies inhibited growth of several tumours in mice and suppressed angiogenesis. Anti-Bv8 treatment also reduced CD11b+Gr1+ cells, both in peripheral blood and in tumours. The effects of anti-Bv8 antibodies were additive to those of anti-Vegf antibodies or cytotoxic chemotherapy. Thus, Bv8 modulates mobilization of CD11b+Gr1+ cells from the bone marrow during tumour development and also promotes angiogenesis locally.


Subject(s)
Gastrointestinal Hormones/metabolism , Myeloid Cells/metabolism , Neoplasms/blood supply , Neovascularization, Pathologic , Neuropeptides/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Gastrointestinal Hormones/antagonists & inhibitors , Gastrointestinal Hormones/immunology , Gene Expression Regulation/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Mice , Mice, Nude , Myeloid Cells/drug effects , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neuropeptides/antagonists & inhibitors , Neuropeptides/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
Nat Biotechnol ; 25(8): 911-20, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17664940

ABSTRACT

Vascular endothelial growth factor (VEGF) is an essential regulator of normal and abnormal blood vessel growth. A monoclonal antibody (mAb) that targets VEGF suppresses tumor growth in murine cancer models and human patients. We investigated cellular and molecular events that mediate refractoriness of tumors to anti-angiogenic therapy. Inherent anti-VEGF refractoriness is associated with infiltration of the tumor tissue by CD11b+Gr1+ myeloid cells. Recruitment of these myeloid cells is also sufficient to confer refractoriness. Combining anti-VEGF treatment with a mAb that targets myeloid cells inhibits growth of refractory tumors more effectively than anti-VEGF alone. Gene expression analysis in CD11b+Gr1+ cells isolated from the bone marrow of mice bearing refractory tumors reveals higher expression of a distinct set of genes known to be implicated in active mobilization and recruitment of myeloid cells. These findings indicate that, in our models, refractoriness to anti-VEGF treatment is determined by the ability of tumors to prime and recruit CD11b+Gr1+ cells.


Subject(s)
Antineoplastic Agents/administration & dosage , CD11b Antigen/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Receptors, Chemokine/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Mice , Mice, Inbred C57BL , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...