Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nat Commun ; 12(1): 6287, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725372

ABSTRACT

Angiopoietin (Angpt)-Tie receptor 2 (Tie2) plays key roles in vascular development and homeostasis as well as pathological vascular remodeling. Therefore, Tie2-agonistic antibody and engineered Angpt1 variants have been developed as potential therapeutics for ischemic and inflammatory vascular diseases. However, their underlying mechanisms for Tie2 clustering and activation remain elusive and the poor manufacturability and stability of Angpt1 variants limit their clinical application. Here, we develop a human Tie2-agonistic antibody (hTAAB), which targets the membrane proximal fibronectin type III domain of Tie2 distinct from the Angpt-binding site. Our Tie2/hTAAB complex structures reveal that hTAAB tethers the preformed Tie2 homodimers into polygonal assemblies through specific binding to Tie2 Fn3 domain. Notably, the polygonal Tie2 clustering induced by hTAAB is critical for Tie2 activation and are resistant to antagonism by Angpt2. Our results provide insight into the molecular mechanism of Tie2 clustering and activation mediated by hTAAB, and the structure-based humanization of hTAAB creates a potential clinical application.


Subject(s)
Antibodies, Monoclonal/chemistry , Receptor, TIE-2/chemistry , Angiopoietin-2/chemistry , Angiopoietin-2/genetics , Angiopoietin-2/immunology , Animals , Antibodies, Monoclonal/immunology , Dimerization , Fibronectins/chemistry , Fibronectins/immunology , Humans , Mice , Mice, Inbred BALB C , Protein Domains , Receptor, TIE-2/agonists , Receptor, TIE-2/genetics , Receptor, TIE-2/immunology , Vascular Remodeling
2.
J Immunol ; 205(8): 2301-2311, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32938724

ABSTRACT

Tie2-expressing monocytes/macrophages (TEMs) are a distinct subset of proangiogenic monocytes selectively recruited to tumors in breast cancer. Because of the hypoxic nature of solid tumors, we investigated if oxygen, via hypoxia-inducible transcription factors HIF-1α and HIF-2α, regulates TEM function in the hypoxic tumor microenvironment. We orthotopically implanted PyMT breast tumor cells into the mammary fat pads of syngeneic LysMcre, HIF-1α fl/fl /LysMcre, or HIF-2α fl/fl /LysMcre mice and evaluated the tumor TEM population. There was no difference in the percentage of tumor macrophages among the mouse groups. In contrast, HIF-1α fl/fl /LysMcre mice had a significantly smaller percentage of tumor TEMs compared with control and HIF-2α fl/fl /LysMcre mice. Proangiogenic TEMs in macrophage HIF-2α-deficient tumors presented significantly more CD31+ microvessel density but exacerbated hypoxia and tissue necrosis. Reduced numbers of proangiogenic TEMs in macrophage HIF-1α-deficient tumors presented significantly less microvessel density but tumor vessels that were more functional as lectin injection revealed more perfusion, and functional electron paramagnetic resonance analysis revealed more oxygen in those tumors. Macrophage HIF-1α-deficient tumors also responded significantly to chemotherapy. These data introduce a previously undescribed and counterintuitive prohypoxia role for proangiogenic TEMs in breast cancer which is, in part, suppressed by HIF-2α.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Macrophages/immunology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Neoplasm Proteins/immunology , Receptor, TIE-2/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/genetics , Oxygen/immunology , Receptor, TIE-2/genetics
3.
Cancer Immunol Res ; 8(2): 268-279, 2020 02.
Article in English | MEDLINE | ID: mdl-31871121

ABSTRACT

Myeloid-derived suppressor cells (MDSC) promote immunosuppression and are a target in the field of immuno-oncology. Accumulation of MDSCs is associated with poor prognosis and resistance to immunotherapy for several cancers. Here, we describe an accumulation of a subset of circulating monocytic MDSCs (M-MDSC) overexpressing TIE2, the receptor for angiopoietin-2 (ANGPT2), in patients with non-small cell lung cancer (NSCLC). Greater numbers of circulating TIE2+ M-MDSCs were detected in patients with NSCLC compared with healthy subjects, and this accumulation correlated with ANGPT2 concentration in blood. The presence of an ANGPT2-rich environment was associated with impairment of preexisting T-cell responses against tumor-associated antigens (TAA) in patients with NSCLC. We demonstrated that ANGPT2 sensitizes TIE2+ M-MDSCs such that these cells suppress TAA-specific T cells. In patients with NSCLC, upregulation of the ANGPT2/TIE2+ M-MDSC signature in blood was associated with a poor prognosis. Our results identify the ANGPT2/TIE2+ M-MDSC axis as a participant in tumor immune evasion that should be taken into account in future cancer immunotherapy.


Subject(s)
Angiopoietin-2/immunology , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/immunology , Myeloid-Derived Suppressor Cells/immunology , Receptor, TIE-2/immunology , T-Lymphocytes/immunology , Tumor Escape , Aged , Aged, 80 and over , Angiopoietin-2/metabolism , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Immune Tolerance , Lung Neoplasms/blood , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Middle Aged , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Staging , Prognosis , Receptor, TIE-2/metabolism , Survival Rate
4.
MAbs ; 10(8): 1260-1268, 2018.
Article in English | MEDLINE | ID: mdl-30199300

ABSTRACT

Ang1 is a soluble ligand to receptor Tie2, and increasing the circulating Ang1 level may improve vascular stabilization under certain disease conditions. Here, we found that the circulating Ang1 level was significantly increased in cynomolgus monkeys treated with non-neutralizing anti-Ang1 antibodies. Improving the antibodies' pharmacokinetic properties by IgG Fc mutations further increased the circulating Ang1 level. However, the mutations decreased the thermal stability of the molecules, which may limit their use as therapeutic antibodies. Nevertheless, we showed that non-neutralizing antibodies may have therapeutic potential by increasing the level of a target molecule in the circulation.


Subject(s)
Angiopoietin-1/immunology , Antibodies, Monoclonal/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Angiopoietin-1/blood , Angiopoietin-1/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Macaca fascicularis , Mutation , Protein Binding , Receptor, TIE-2/genetics , Receptor, TIE-2/immunology , Receptor, TIE-2/metabolism
5.
Biomaterials ; 51: 119-128, 2015 May.
Article in English | MEDLINE | ID: mdl-25771003

ABSTRACT

Angiopoietin-1 (Ang1) and its endothelium-specific receptor, tyrosine kinase with Ig and epidermal growth factor homology domain 2 (Tie2), play critical roles in vascular development. Although the Ang1/Tie2 system has been considered a promising target for therapeutic neovascularization, several imitations of large-scale production have hampered the development of recombinant Ang1 for therapeutics. In this study, we produced a fully human agonistic antibody against Tie2, designated 1-4h, and tested the applicability of 1-4h as an alternative to native Ang1 in therapeutic angiogenesis. 1-4h significantly enhanced the phosphorylation of Tie2 in a dose- and time-dependent manner in human Tie2-expressing HEK293 cells and human umbilical vein endothelial cells. Moreover, 1-4h induced the activation of Tie2-mediated intracellular signaling such as AKT, eNOS, MAPK, and Focal Adhesion Kinase p125(FAK). In addition, 1-4h increased the chemotactic motility and capillary-like tube formation of endothelial cells in vitro and enhanced the survival of serum-deprived endothelial cells. Taken together, our data clearly suggest that a human Tie2 agonistic antibody is a potentially useful therapeutic approach for the treatment of several ischemic diseases including delayed-wound healing and ischemic heart and limb diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Neovascularization, Physiologic/drug effects , Receptor, TIE-2/immunology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Survival/drug effects , HEK293 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Space/metabolism , Mice , NIH 3T3 Cells , Peptide Library , Protein Binding/drug effects , Signal Transduction/drug effects , Single-Chain Antibodies/metabolism
6.
Mol Cells ; 36(5): 432-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24158612

ABSTRACT

Tumor-associated macrophages (TAMs) accumulate in various cancers and promote tumor angiogenesis and metastasis, and thus may be ideal targets for the clinical diagnosis of tumor metastasis with high specificity. However, there are few specific markers to distinguish between TAMs and normal or inflammatory macrophages. Here, we show that TAMs localize in green fluorescent protein-labeled tumors of metastatic lymph nodes (MLNs) from B16F1 melanoma cells but not in necrotic tumor regions, suggesting that TAMs may promote the growth of tumor cells and the progression of tumor metastasis. Furthermore, we isolated pure populations of TAMs from MLNs and characterized their gene expression signatures compared to peritoneal macrophages (PMs), and found that TAMs significantly overexpress immunosuppressive cytokines such as IL-4, IL-10, and TGF-ß as well as proangiogenic factors such as VEGF, TIE2, and CD31. Notably, immunological analysis revealed that TIE2(+)/CD31(+) macrophages constitute the predominant population of TAMs that infiltrate MLNs, distinct from tissue or inflammatory macrophages. Importantly, these TIE2(+)/CD31(+) macrophages also heavily infiltrated MLNs from human breast cancer biopsies but not reactive hyperplastic LNs. Thus, TIE2(+)/ CD31(+) macrophages may be a unique histopathological biomarker for detecting metastasis in clinical diagnosis, and a novel and promising target for TAM-specific cancer therapy.


Subject(s)
Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Macrophages/physiology , Melanoma, Experimental/secondary , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Receptor, TIE-2/analysis , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/immunology , Lymphatic Metastasis/immunology , Macrophages/immunology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Receptor, TIE-2/immunology , Tumor Microenvironment
7.
J Immunol Methods ; 391(1-2): 60-71, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23454004

ABSTRACT

Phage display antibody libraries have a proven track record for the discovery of therapeutic human antibodies, increasing the demand for large and diverse phage antibody libraries for the discovery of new therapeutics. We have constructed naïve antibody phage display libraries in both Fab and scFv formats, with each library having more than 250 billion clones that encompass the human antibody repertoire. These libraries show high fidelity in open reading frame and expression percentages, and their V-gene family distribution, VH-CDR3 length and amino acid usage mirror the natural diversity of human antibodies. Both the Fab and scFv libraries show robust sequence diversity in target-specific binders and differential V-gene usage for each target tested, supporting the use of libraries that utilize multiple display formats and V-gene utilization to maximize antibody-binding diversity. For each of the targets, clones with picomolar affinities were identified from at least one of the libraries and for the two targets assessed for activity, functional antibodies were identified from both libraries.


Subject(s)
Cell Surface Display Techniques , Immunoglobulin Fab Fragments/immunology , Peptide Library , Receptor, Insulin/immunology , Receptor, TIE-2/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antibody Affinity , Antibody Specificity , CHO Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/genetics , Mitogen-Activated Protein Kinases/metabolism , Open Reading Frames , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/genetics , Receptor, TIE-2/genetics , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Transfection
8.
PLoS One ; 8(2): e54923, 2013.
Article in English | MEDLINE | ID: mdl-23405099

ABSTRACT

There is increasing experimental evidence for an important role of Angiopoietin-2 (Ang-2) in tumor angiogenesis and progression. In addition, Ang-2 is up-regulated in many cancer types and correlated with poor prognosis. To investigate the functional role of Ang-2 inhibition in tumor development and progression, we generated novel fully human antibodies that neutralize specifically the binding of Ang-2 to its receptor Tie2. The selected antibodies LC06 and LC08 recognize both rodent and human Ang-2 with high affinity, but LC06 shows a higher selectivity for Ang-2 over Ang-1 compared to LC08 which can be considered an Ang-2/Ang-1 cross-reactive antibody. Our data demonstrate that Ang-2 blockade results in potent tumor growth inhibition and pronounced tumor necrosis in subcutaneous and orthotopic tumor models. These effects are attended with a reduction of intratumoral microvessel density and tumor vessels characterized by fewer branches and increased pericyte coverage. Furthermore, anti-Ang-2 treatment strongly inhibits the dissemination of tumor cells to the lungs. Interestingly, in contrast to the Ang-2/Ang-1 cross-reactive antibody LC08 that leads to a regression of physiological vessels in the mouse trachea, the inhibition with the selective anti-Ang-2 antibody LC06 appears to be largely restricted to tumor vasculature without obvious effects on normal vasculature. Taken together, these data provide strong evidence for the selective Ang-2 antibody LC06 as promising new therapeutic agent for the treatment of various cancers.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Angiopoietin-1/immunology , Animals , Antibodies, Neutralizing/immunology , Antibody Specificity , Antineoplastic Agents/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cornea/drug effects , Cornea/immunology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Microvessels/drug effects , Microvessels/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Phosphorylation , Random Allocation , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/immunology , Xenograft Model Antitumor Assays
9.
PLoS One ; 6(8): e23540, 2011.
Article in English | MEDLINE | ID: mdl-21858161

ABSTRACT

BACKGROUND: Angiogenesis is a critical early event in inflammatory arthritis, facilitating leukocyte migration into the synovium resulting in invasion and destruction of articular cartilage and bone. This study investigates the effect of TLR2 on angiogenesis, EC adhesion and invasion using microvascular endothelial cells and RA whole tissue synovial explants ex-vivo. METHODS: Microvascular endothelial cells (HMVEC) and RA synovial explants ex vivo were cultured with the TLR2 ligand, Pam3CSK4 (1 µg/ml). Angiopoietin 2 (Ang2), Tie2 and TLR2 expression in RA synovial tissue was assessed by immunohistology. HMVEC tube formation was assessed using Matrigel matrix assays. Ang2 was measured by ELISA. ICAM-1 cell surface expression was assessed by flow cytometry. Cell migration was assessed by wound repair scratch assays. ECM invasion, MMP-2 and -9 expression were assessed using transwell invasion chambers and zymography. To examine if the angiopoietin/Tie2 signalling pathway mediates TLR2 induced EC tube formation, invasion and migration assays were performed in the presence of a specific neutralising anti-Tie2mAb (10 ug/ml) and matched IgG isotype control Ab (10 ug/ml). RESULTS: Ang2 and Tie2 were localised to RA synovial blood vessels, and TLR2 was localised to RA synovial blood vessels, sub-lining infiltrates and the lining layer. Pam3CSK4 significantly increased angiogenic tube formation (p<0.05), and upregulated Ang2 production in HMVEC (p<0.05) and RA synovial explants (p<0.05). Pam3CSK4 induced cell surface expression of ICAM-1, from basal level of 149±54 (MFI) to 617±103 (p<0.01). TLR-2 activation induced an 8.8±2.8 fold increase in cell invasion compared to control (p<0.05). Pam3CSK4 also induced HMVEC cell migration and induced MMP-2 and -9 from RA synovial explants. Neutralisation of the Ang2 receptor, Tie2 significantly inhibited Pam3CSK4-induced EC tube formation and invasion (p<0.05). CONCLUSION: TLR2 activation promotes angiogenesis, cell adhesion and invasion, effects that are in part mediated through the Tie2 signalling pathway, key mechanisms involved in the pathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Neovascularization, Physiologic/physiology , Receptor, TIE-2/physiology , Signal Transduction/physiology , Toll-Like Receptor 2/physiology , Angiopoietin-2/metabolism , Antibodies/immunology , Antibodies/pharmacology , Arthritis, Rheumatoid/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Lipopeptides/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Physiologic/drug effects , Receptor, TIE-2/immunology , Receptor, TIE-2/metabolism , Signal Transduction/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tissue Culture Techniques , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/metabolism
10.
Cancer Res ; 70(24): 10150-60, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159637

ABSTRACT

The inhibition of VEGF signaling with antibodies or small molecules achieves clinical benefits in diverse solid malignancies. Nonetheless, therapeutic effects are usually not sustained, and most patients eventually succumb to progressive disease, indicating that antiangiogenic strategies require additional optimization. Vaccination with lethally irradiated, autologous tumor cells engineered to secrete granulocyte-macrophage colony stimulating factor (GM-CSF) and antibody blockade of cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) trigger a tumor vasculopathy in some long-term responding subjects. These reactions are characterized by disrupted tumor blood vessels in association with lymphocyte and granulocyte infiltrates and zonal areas of ischemic tumor necrosis. However, the mechanisms underlying this immune-mediated destruction of the tumor vasculature remain to be clarified. Here, we show that GM-CSF-secreting tumor cell vaccines and CTLA-4 blockade elicit a functionally important humoral reaction against multiple angiogenic cytokines. Antibodies to angiopoietin-1 and angiopoietin-2 block Tie-2 binding, downstream signaling, endothelial cell tube formation, and macrophage chemotaxis. Antibodies to macrophage inhibitory factor (MIF) attenuate macrophage Tie-2 expression and matrix metalloproteinase-9 (MMP-9) production. Together, these results delineate an immunotherapy-induced host response that broadly targets the angiogenic network in the tumor microenvironment.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Cancer Vaccines/immunology , Immunotherapy, Active/methods , Neoplasms/blood supply , Neoplasms/therapy , Angiopoietin-1/immunology , Angiopoietin-2/immunology , Animals , Antibodies, Neoplasm/immunology , Antibody Formation , Antigens, CD/immunology , CTLA-4 Antigen , Cancer Vaccines/administration & dosage , Gene Library , Humans , Immunity, Humoral , Melanoma/immunology , Melanoma/therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/therapy , Receptor, TIE-2/immunology , Vascular Endothelial Growth Factor A/immunology
11.
J Immunol Methods ; 355(1-2): 21-8, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20188106

ABSTRACT

A well-designed anti-drug antibody (ADA) immunoassay is critical for appropriately monitoring the immunogenicity profile of a therapeutic protein during its development. AMG 386 is a peptide-Fc fusion protein that inhibits angiogenesis by preventing the interaction of angiopoietins with the Tie2 receptor. In bridging immunoassays for ADA, interference by the drug target, present in the assay sample, can result in false positive antibody detection. We used a statistical design-of-experiments approach to identify angiopoietin interference in bridging immunoassays of anti-AMG 386 antibodies. We also demonstrated that a high-affinity monoclonal antibody, directed against an epitope on angiopoietin that competes with AMG 386 binding, could inhibit the angiopoietin interference while preserving the detection of ADA. This report describes the development and validation of methodologies for evaluating and addressing drug target interference in bioanalytical assays that involve interactions between drug, ADA, immune complexes, and drug target.


Subject(s)
Antibodies, Monoclonal/chemistry , Drug Interactions , Immunoglobulin Fc Fragments/analysis , Neoplasms/blood , Neovascularization, Pathologic/blood , Peptides/analysis , Angiopoietins/blood , Angiopoietins/genetics , Angiopoietins/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Biological Assay , Clinical Trials as Topic , Female , Humans , Immunoassay/methods , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Male , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Peptides/genetics , Peptides/immunology , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/genetics , Receptor, TIE-2/immunology , Receptor, TIE-2/metabolism , Sensitivity and Specificity
12.
Rom J Morphol Embryol ; 51(1): 81-4, 2010.
Article in English | MEDLINE | ID: mdl-20191124

ABSTRACT

Tie2 is a member of receptor tyrosine kinases family, involved in vasculogenesis and angiogenesis. Its main role is to stabilize, maintain, and facilitate the structural adaptation of the vasculature, during embryo development, and adult wound healing, or tumor development. Tissues from human embryos found in different stages of development (5 and 7-week-old), were investigated for Tie2 expression. The reaction was positive, with maximum intensity in the vascular cords, found in the mesenchymal tissue, and in the connective tissue around the primitive spinal cord. In the 7-week-old embryo, the reaction was negative in large blood vessels, and it was heterogeneous in those showing bridging phenomenon. In conclusion, during the first two months of human embryo development, we have concurrent vasculogenesis, angiogenesis, and blood vessel maturation and stabilization.


Subject(s)
Embryo, Mammalian/metabolism , Fetus/metabolism , Receptor, TIE-2/metabolism , Adult , Blood Vessels/metabolism , Gestational Age , Humans , Mesoderm/metabolism , Receptor, TIE-2/immunology
13.
Expert Opin Investig Drugs ; 19(2): 169-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20050812

ABSTRACT

BACKGROUND: The Tie-2 receptor can bind its agonistic ligand Angiopoietin-1 (Ang-1) and the potential antagonist Ang-2. Tie-2 can be expressed both by primary human acute myeloid leukaemia (AML) cells and endothelial cells, and Tie-2-blocking antibodies are now being evaluated in clinical trials for cancer treatment. DESIGN AND METHODS: We investigated the effects of Tie-2-blocking antibodies, exogenous Ang-2 and pharmacological agents on AML cell proliferation and the release of angioregulatory mediators. RESULTS: Tie-2-blocking antibodies had a growth inhibitory effect on human AML cells co-cultured with microvascular endothelial cells, but this inhibition was not observed when leukaemic cells were co-cultured with fibroblasts or osteoblasts. AML cell viability in co-cultures was not altered by anti-Tie-2. Furthermore, anti-Tie-2 decreased hepatocyte growth factor (HGF) levels and increased CXCL8 levels in co-cultures, whereas the levels of endocan (a proteoglycan released by endothelial cells) were not altered. The only significant effects of exogenous Ang-2 were decreased levels of HGF and endocan. Constitutive AML cell release of agonistic Ang-1 was decreased by the proteasomal inhibitor bortezomib and the specific IkappaB-kinase/NFkappaB inhibitor BMS-345541. CONCLUSION: We conclude that various strategies for inhibition of Tie-2-mediated signalling should be considered in AML therapy, possibly in combination with other antiangiogenic strategies.


Subject(s)
Angiopoietin-1/metabolism , Angiopoietin-2/pharmacology , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Receptor Cross-Talk/drug effects , Receptor, TIE-2/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Angiopoietin-2/metabolism , Antibodies/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Female , Fibroblasts/drug effects , Hepatocyte Growth Factor/metabolism , Humans , Imidazoles/pharmacology , Interleukin-8/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Osteoblasts/drug effects , Proteoglycans/metabolism , Pyrazines/pharmacology , Quinoxalines/pharmacology , Receptor, TIE-2/immunology , Signal Transduction/drug effects
14.
Atherosclerosis ; 204(2): 365-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19022447

ABSTRACT

BACKGROUND: TIE2(+) cells play a crucial role in processes that are involved in atherosclerosis, such as angiogenesis. Therefore, the specific deletion of TIE2(+) cells by means of DNA vaccination may affect atherosclerosis. METHODS: Cellular immunity against cells that overexpress TIE2 was established in LDLr(-/-) mice by a novel oral DNA vaccination technique, in which an attenuated Salmonella typhimurium strain was used as a carrier for plasmid pcDNA3.1 encoding TIE2. After three oral vaccinations with 2-week time intervals LDLr(-/-) mice were put on a Western type diet and atherosclerosis was induced. RESULTS: Eight weeks after vaccination FACS analysis of circulating peripheral blood mononuclear cells (PBMCs) revealed a significant decrease (33%, p<0.05) in TIE2(+) cells upon vaccination against TIE2, indicating the successful induction of cellular immunity following vaccination against TIE2. Six weeks after collar placement vaccination against TIE2 resulted in significantly decreased carotid atherosclerosis, as indicated by 30% (p<0.05) reduced intima area and 27% (p<0.05) reduced intima/lumen ratios. Furthermore, atherosclerosis was attenuated in the aortic root by 42% (p<0.05), further underlining the anti-atherosclerotic effect of vaccination against TIE2. Adventitial angiogenesis was reduced by 61% (p<0.05) upon vaccination against TIE2 providing a mechanism via which vaccination against TIE2 inhibits lesion formation. Histochemical analysis of the atherosclerotic lesion composition revealed a 1.6-fold (carotid artery, p<0.05) and 1.9-fold (aortic root, p<0.05) increase in collagen content upon vaccination against TIE2, indicating a more stable plaque phenotype. CONCLUSIONS: We demonstrate that vaccination against TIE2 induces cellular immunity against cells that overexpress TIE2 and results in smaller atherosclerotic lesions with a more stable phenotype. Therefore, vaccination strategies that target cells that contribute to atherosclerosis, may be of potential use in the development of novel treatments of atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Immunity, Cellular , Leukocytes, Mononuclear/immunology , Receptor, TIE-2/immunology , Vaccines, DNA/administration & dosage , Administration, Oral , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Collagen/metabolism , Disease Models, Animal , Female , Genetic Vectors , Immunization Schedule , Mice , Mice, Knockout , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Phenotype , Receptor, TIE-2/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Salmonella typhimurium/genetics , Vaccines, Attenuated/administration & dosage
15.
Prostate ; 67(15): 1664-76, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17854058

ABSTRACT

BACKGROUND: Castration results in a major involution of the normal prostate gland. This process is initiated by effects in the prostate stroma and vasculature. Castration-induced regression of androgen sensitive prostate tumors is however less prominent and hypothetically this could be related to a limited stromal/vascular response. We therefore used animal tumor models to explore the importance of stroma and vascular effects, and if castration effects could be enhanced by a simultaneous therapy targeting the tumor stroma. METHODS: Using rats with Dunning PAP and H tumors, stereological methods, immunohistochemistry, and Western blotting, we studied the tumor response 7 and 28 days after castration and after the addition of stroma targeted therapies. RESULTS: In the normal ventral prostate (VP) nuclear androgen receptors (AR) were rapidly downregulated after castration. In contrast, the Dunning tumors downregulated the AR in the cancerous epithelium, but not in the surrounding stroma. Vascular regulators such as the angiopoietins, tie 2, and PDGF-Rbeta were not decreased in the stroma after castration, as observed in the VP, creating an environment that prevents vascular involution. When a tumor stroma targeted therapy inhibiting the tie 2 receptor and the PDGF-Rbeta simultaneously was added to castration it resulted in a decreased vascular density, increased tumor cell apoptosis and decreased tumor growth compared to castration alone. CONCLUSIONS: The stroma in highly differentiated androgen sensitive Dunning tumors is apparently androgen insensitive. If this unresponsive stroma is targeted the effects of castration can be enhanced.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Orchiectomy , Piperazines/pharmacology , Prostatic Neoplasms/drug therapy , Pyrimidines/pharmacology , Receptor, TIE-2/metabolism , Stromal Cells/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Androgen Receptor Antagonists , Angiopoietins/antagonists & inhibitors , Angiopoietins/metabolism , Animals , Benzamides , Disease Models, Animal , Down-Regulation , Drug Therapy, Combination , Imatinib Mesylate , Immunoglobulin Fc Fragments/pharmacology , Male , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Prostate/blood supply , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Rats , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/immunology , Receptors, Androgen/metabolism , Recombinant Fusion Proteins/pharmacology , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
16.
Methods Enzymol ; 420: 255-64, 2006.
Article in English | MEDLINE | ID: mdl-17161700

ABSTRACT

Hematopoietic stem cells (HSCs) primarily reside in bone marrow, are defined by their ability to maintain blood homeostasis, and replenish themselves through self-renewal. Although HSC purification schemes vary from laboratory to laboratory, the resulting cell populations are similar, if not the same. This chapter will discuss different enrichment methods for HSCs and provide a detailed protocol for staining HSC with Hoechst 33342 for the side population (SP).


Subject(s)
Antigens, Differentiation/analysis , Benzimidazoles , Cell Separation/methods , Flow Cytometry/methods , Hematopoietic Stem Cells/cytology , Membrane Proteins/analysis , Animals , Antigens, CD/analysis , Antigens, CD/immunology , Antigens, Differentiation/biosynthesis , Endoglin , Fluorescent Dyes , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/biosynthesis , Receptor, TIE-2/analysis , Receptor, TIE-2/immunology , Receptors, Cell Surface/analysis , Receptors, Cell Surface/immunology , Staining and Labeling
17.
Clin Cancer Res ; 12(6): 1813-9, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16551866

ABSTRACT

PURPOSE: Tie-2 is an endothelium-specific receptor tyrosine kinase known to play a key role in tumor angiogenesis. The present study explores the feasibility of immunotherapy of tumors by using a protein vaccine based on chicken Tie-2 as a model antigen to break the immune tolerance against Tie-2 in a cross-reaction between the xenogeneic homologous and self-Tie-2. EXPERIMENTAL DESIGN AND RESULTS: In this study, a chicken homologous Tie-2 protein vaccine (chTie-2) and a corresponding mouse Tie-2 vaccine as a control were prepared and the antitumor effect of these vaccines was tested in two tumor models (murine B16F10 melanoma and murine H22 hepatoma). Immunotherapy with chTie-2 was found effective in two tumor models. Autoantibodies against mouse Tie-2 were detected in sera of mice immunized with chTie-2 through Western blot analysis and ELISA assay. Anti-Tie-2 antibody-producing B cells were detectable by ELISPOT. Histologic examination revealed that autoantibodies were deposited on the endothelial cells of tumor tissues. Purified immunoglobulins from chTie-2-immunized mice could induce the apoptosis of human umbilical vein endothelial cells in vitro. Importantly, adoptive transfer of purified immunoglobulins led to antitumor effect in vivo; apparently, angiogenesis was significantly inhibited in these tumors. Furthermore, the antitumor activity and production of autoantibodies could be abrogated by depletion of CD4+ T lymphocytes. CONCLUSIONS: Our findings may provide a vaccine strategy for cancer therapy and show the potential utilization of interference with Tie-2 pathway.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasms, Experimental/therapy , Receptor, TIE-2/immunology , Animals , Apoptosis/drug effects , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Line , Cell Line, Tumor , Chickens , Humans , Immunoglobulins/immunology , Immunoglobulins/pharmacology , Immunoglobulins/therapeutic use , Immunohistochemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Survival Analysis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Time Factors , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology , Vaccines, Subunit/therapeutic use
18.
Cancer Immunol Immunother ; 55(8): 1004-10, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16408213

ABSTRACT

A potential target for a cancer vaccine would be receptors, such as Tie-2 which are over expressed on tumour endothelium. Using computer aided motif predictions for possible HLA class I epitopes, we have identified peptides from Tie-2 that should bind with a range of affinities to HLA-A*0201. No direct correlation between predicted values and actual binding affinities was observed. Although, the programs did produce a number of false positives, two epitopes were predicted that bound with relatively high affinity when compared with an influenza peptide. We have previously identified a Tie-2 epitope and shown that it was only immunogenic when we substituted preferred amino acids at key anchor residues to increase binding affinity. In this study we used a similar approach to generate modified epitopes. When HLA-A2 transgenic mice were immunised with peptides, CTL killing of the target cells was only achieved when the wild type epitope was presented at moderate levels. Moreover, the efficiency of immunisation was increased when we linked CD4 epitopes to CD8 epitopes. Caution should therefore be employed in the use of both reverse immunology and anchor modification of CTL epitopes in the identification of CTL epitopes for cancer vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/metabolism , Immunologic Techniques , Receptor, TIE-2/immunology , Algorithms , Animals , Base Sequence , Binding Sites , Mice , Mice, Transgenic , Molecular Sequence Data , T-Lymphocytes, Cytotoxic/immunology
19.
Proc Natl Acad Sci U S A ; 102(23): 8293-8, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15928093

ABSTRACT

The endothelial cell receptor-tyrosine kinases, VEGF receptor 2 (VEGF-R2) and Tie-2, and their ligands, vascular endothelial growth factor (VEGF) and angiopoietins 1 and 2, respectively, play key roles in tumor angiogenesis. Several studies suggest that the VEGF receptor pathway and the Tie-2 pathway are independent and essential mediators of angiogenesis, leading to the hypothesis that simultaneous interference with both pathways should result in additive effects on tumor growth. In this study, a human melanoma xenograft model (M21) was used to analyze the effects of simultaneous intradiabody depletion of vascular endothelial growth receptor-R2 and Tie-2 on tumor angiogenesis and tumor xenograft growth. The intradiabodies were expressed from recombinant adenovirus delivered through subtumoral injection. Blockade of both VEGF-R2 and Tie-2 pathways simultaneously or the VEGF receptor pathway alone resulted in a significant inhibition of tumor growth and tumor angiogenesis (92.2% and 74.4%, respectively). In addition, immunohistochemical staining of intradiabody-treated tumors demonstrated a decreased number of tumor-associated blood vessels versus control treatment. Previous studies with intrabodies had demonstrated that the Tie-2 receptor pathway was essential for tumor growth. The simultaneous blockade of the VEGF and Tie-2 pathways resulted in effective inhibition of tumor growth and demonstrated the potential of simultaneous targeting of multiple pathways as a therapeutic strategy.


Subject(s)
Immunoglobulin Fab Fragments/immunology , Melanoma/blood supply , Melanoma/pathology , Neovascularization, Pathologic , Receptor, TIE-2/deficiency , Receptors, Vascular Endothelial Growth Factor/deficiency , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/pharmacology , Melanoma/immunology , Melanoma/therapy , Mice , Neoplasm Transplantation , Phenotype , Rabbits , Receptor, TIE-2/immunology , Receptor, TIE-2/metabolism , Receptors, Vascular Endothelial Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Clin Immunol ; 115(1): 93-101, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15870027

ABSTRACT

Our previous studies of gene expression profiling during collagen-induced arthritis (CIA) indicated that the putative angiogenic factor Angptl4 was one of the most highly expressed mRNAs early in disease. To investigate the potential involvement of Angptl4 in CIA pathogenesis, Angptl4 protein levels were assessed at early stages of disease and its cellular sources were determined. In addition, the functional effects of mouse Angptl4 on endothelial cells were assessed. Angptl4 protein levels were higher in arthritic joints as compared to normal joints. In situ hybridization localized Angptl4 mRNA to stromal fibroblast-like cells within the inflamed synovium. Temporal expression of Angptl4 mRNA during CIA was similar to that of key angiogenic factors, including structurally related angiopoietin 1. Recombinant mouse Angptl4 promoted endothelial cell survival and formation of tubule-like structures. These functional effects of Angptl4, combined with very high expression at early stages of CIA, suggest a role for Angptl4 in angiogenesis in arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Blood Proteins/immunology , Neovascularization, Pathologic/immunology , Angiopoietin-1/immunology , Angiopoietin-Like Protein 4 , Angiopoietins , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Blood Proteins/biosynthesis , Blood Proteins/genetics , Blotting, Western , Collagen , Endothelial Cells , Humans , In Situ Hybridization , Interleukin-1 Receptor-Like 1 Protein , Male , Membrane Proteins/immunology , Mice , Mice, Inbred DBA , Oligonucleotide Array Sequence Analysis , Platelet Endothelial Cell Adhesion Molecule-1/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, TIE-2/immunology , Receptors, Interleukin , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL
...