Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
2.
Parasite Immunol ; 35(3-4): 109-119, 2013.
Article in English | MEDLINE | ID: mdl-23163958

ABSTRACT

Leishmania amazonensis parasites can cause diverse forms of leishmaniasis in humans and persistent lesions in most inbred strains of mice. In both cases, the infection is characterized by a marked immunosuppression of the host. We previously showed that amastigote forms of the parasite make use of surface-exposed phosphatidylserine (PS) molecules to infect host cells and promote alternative macrophage activation, leading to uncontrolled intracellular proliferation of the parasites. In this study, we demonstrated that treatment of infected mice with a PS-targeting monoclonal antibody ameliorated parasite loads and lesion development, which correlated with increased proliferative responses by lymphocytes. In addition, we observed an enhanced dendritic cell (DC) activation and antigen presentation in vitro. Our data imply that the recognition of PS exposed on the surface of amastigotes plays a role in down-modulating DC functions, in a matter similar to that of apoptotic cell clearance. This study provides new information regarding the mechanism of immune suppression in Leishmania infection.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/parasitology , Leishmania mexicana/immunology , Phosphatidylserines/immunology , Animals , Disease Models, Animal , Female , Immune Evasion , Immune Tolerance , Leishmania mexicana/chemistry , Mice , Phosphatidylserines/analysis
5.
FEBS Lett ; 581(4): 702-6, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17274989

ABSTRACT

Oxidative stress is one of the factor contributing to blood brain barrier degeneration. This phenomenon is observed during pathological conditions such as Alzheimer's disease or cerebral amyloid angiopathy in which brain haemorrhages are very frequent. Both diseases are characterized by beta amyloid peptide deposition either in neurons or in vessels. Oxidative stress leads to impairment of mitochondrial functions and apoptotic cell death subsequent to caspases activation. In this paper we demonstrate that BH4 domain of Bcl-xl administrated to endothelial cells as the conjugated form with TAT peptide, reverts Abeta-induced apoptotic cell death by activating a survival programme which is Akt/endothelial nitric oxide synthase dependent.


Subject(s)
Amyloid beta-Peptides/toxicity , Endothelium, Vascular/drug effects , Gene Products, tat/pharmacology , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cattle , Cell Survival/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Protein Structure, Tertiary/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism
6.
Appl Radiat Isot ; 63(3): 343-51, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15955705

ABSTRACT

Radioarsenic labelled radiopharmaceuticals could be a valuable asset to Positron Emission Tomography (PET). In particular, the long half-lives of (72)As (T(1/2)=26 h) and (74)As (T(1/2)=17.8 d) allow to investigate slow physiological or metabolical processes, like the enrichment and distribution of antibodies in tumor tissue. This work describes the direct production of no-carrier-added (nca) arsenic isotopes *As, with *=71, 72, 73, 74 or 77, the reaction to [*As]AsI(3) and its radiochemical separation from the irradiated solid germanium oxide via polystyrene-based solid-phase extraction. The germanium oxide target, irradiated at a cyclotron or a nuclear reactor, is dissolved in concentrated HF and Ge is separated almost quantitatively (99.97%) as [GeF(6)](2-). [*As]AsI(3) is formed by addition of potassium iodide. The radiochemical separation yield for arsenic is >90%. [*As]AsI(3) is a versatile radioarsenic labelling synthon.


Subject(s)
Arsenic/isolation & purification , Germanium/chemistry , Radioisotopes/isolation & purification , Radiopharmaceuticals/chemical synthesis , Half-Life , Positron-Emission Tomography/methods
7.
J Vasc Res ; 41(2): 202-7, 2004.
Article in English | MEDLINE | ID: mdl-15031604

ABSTRACT

BACKGROUND: Vascular endothelium undergoes apoptosis when exposed to reactive oxygen species (ROS), including hydrogen peroxide and superoxide radicals. ROS are believed to be the cause of damage to small vessels during ischemia-reperfusion injury and of arterial damage during atherosclerosis. Hydrogen peroxide-induced apoptosis is mediated through the inhibition of Bcl-xl activity and caspase-3 and caspase-9 activation. The BH4 domain of the Bcl-2 family members is responsible for their antiapoptotic activity. The BH4 domains of Bcl-2 and Bcl-xl inhibit cytochrome c release and the loss of mitochondrial membrane potential. METHODS AND RESULTS: The purpose of this project was to study the antiapoptotic effect of cell-permeant derivative of Bcl-2 (BH4 peptide) on endothelial cells exposed to stress conditions. BH4 peptide was conjugated to the cell-permeable peptide TAT and was applied to endothelial cells under conditions of serum starvation and hydrogen peroxide treatment. TAT-BH4 reduced caspase-3 activity and prevented apoptotic cell death. CONCLUSION: Our results indicate that TAT-BH4 peptide can protect endothelial cells from ROS-induced apoptosis.


Subject(s)
Apoptosis/physiology , Coronary Vessels/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Aorta/cytology , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Humans , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , Swine , Umbilical Veins/cytology
8.
J Thromb Haemost ; 2(3): 459-67, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15009464

ABSTRACT

The procoagulant activity (PCA) of four T-lymphoblastoid cell lines (CEM-CCRF, Jurkat, Molt-4 and A3.01) at different stages of differentiation has been characterized and compared with that of a monocytoid cell line (THP-1). Four assay systems were employed; the activated partial thromboplastin time (APTT); prothrombin time/tissue factor (TF) activity; a purified factor (F)Xa generation system and cancer procoagulant. High levels of TF activity were seen only with the monocytic cells. However the more differentiated of the T-lymphoblastoid cells (Molt-4 and A3.01) were more active than monocytic cells in supporting FXa generation. This pattern was not repeated for the APTT assay, which was related to cell-surface TF activity, since it was partially inhibited by antiTF antibody. Annexin V totally inhibited the activity observed in all three assay systems, indicating that the PCA of T-lymphoblastoid cells is primarily due to expression of negatively charged phospholipids. However, antiphosphatidylserine antibody even at a high concentration gave only partial inhibition of the activity observed in the APTT and FXa generation systems for the cells compared with almost total inhibition for the phospholipid standard, suggesting either that cellular phosphatidylserine (PS) is less accessible to the antibody, or that PS is not the sole negatively charged phospholipid responsible for this activity. Flow cytometry studies using propidium iodide and annexin V showed that the PCA, although linked to PS exposure, was not the result of apoptosis.


Subject(s)
Partial Thromboplastin Time , Prothrombin Time , T-Lymphocytes/physiology , Thromboplastin/physiology , Apoptosis , Cell Differentiation , Cell Line , Cell Membrane/physiology , Humans , Jurkat Cells , T-Lymphocytes/cytology
10.
Cancer Res ; 61(23): 8569-77, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731444

ABSTRACT

The gene for the major angiogenic factor, vascular endothelial growth factor (VEGF), encodes several spliced isoforms. We reported previously that overexpression of two VEGF isoforms, VEGF(121) and VEGF(165), by human glioma U87 MG cells induced tumor-associated intracerebral hemorrhage, whereas expression of a third form, VEGF(189), did not cause vessel rupture. Here, we test whether these VEGF isoforms have distinct activities for enhancing vascularization and growth of gliomas in mice. U87 MG cells that overexpressed VEGF(165) or VEGF(189) grew more rapidly than the parental cells in both s.c. and intracranial (i.c.) locations. However, cells that overexpressed VEGF(121) only showed enhancement of i.c. tumor growth but had a minimal effect on s.c. glioma progression. At both anatomical sties, VEGF(165) and VEGF(189) strongly augmented neovascularization, whereas VEGF(121) only increased vessel density in brain tumors. In each type of glioma, expression of VEGF receptors -1 and -2 largely phenocopied the tumor vasculature, because increased VEGF/VEGF receptor-activated microvessel densities were strongly correlated with the angiogenicity and tumorigenicity elicited by the VEGF isoforms at both anatomical sites. One notable difference between the sites was the expression of vitronectin, a prototypic ligand of alpha(v)beta(3) and alpha(v)beta(5) integrins, detected in i.c. but not in s.c., gliomas. Endothelial cell migration stimulated by VEGF(121) was potentiated by vitronectin to a greater extent than that stimulated by VEGF(165). This data demonstrates that VEGF isoforms have distinct activities at different anatomical sites and suggest that the microenvironment of different tissues affects the function of VEGF isoforms.


Subject(s)
Brain Neoplasms/blood supply , Endothelial Growth Factors/physiology , Glioma/blood supply , Lymphokines/physiology , Neovascularization, Pathologic/metabolism , Animals , Brain Neoplasms/metabolism , Cell Movement/drug effects , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glioma/metabolism , Humans , Lymphokines/biosynthesis , Lymphokines/pharmacology , Neovascularization, Pathologic/pathology , Protein Isoforms , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vitronectin/pharmacology
11.
Cancer ; 92(10): 2569-77, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11745191

ABSTRACT

BACKGROUND: Vascular endothelial growth factor (VEGF) is an important endothelial cell mitogen associated with increased angiogenesis and aggressive tumor behavior. Its stimulating effect on endothelial cells basically is dependent on the presence of specific VEGF receptors, such as the flk-1(KDR) receptor. This study investigates the roles of VEGF and of a functionally intact angiogenic pathway, "VEGF/flk-1(KDR)," in patients with endometrial carcinoma and their significance in prognosis and therapy. METHODS: A series of 121 endometrial carcinomas were studied. The expression of VEGF by endometrial tumor cells was assessed using the monoclonal antibody (MoAb) VG1. VEGF/KDR complexes on tumor endothelium or activated microvessel density (aMVD) were identified using the MoAb 11B5. In addition, the standard microvessel density (sMVD) was assessed with anti-CD31. In all tumors, the alkaline phosphatase/antialkaline phosphatase technique was employed. A Fisher exact test or an unpaired, two-tailed t test was used for testing correlations between categoric tumor variables, whereas a log-rank test was used to determine statistical differences between life tables. A Cox proportional hazards model was used to assess the effect of tumor variables on overall survival. RESULTS: Cytoplasmic VEGF expression in > 50% of tumor cells was associated significantly with aMVD (P < 0.0001) and with sMVD (P < 0.003). In univariate survival analysis, VEGF (P = 0.0002), aMVD (P = 0.001), and sMVD (P = 0.0009) were significant prognostic variables. Equally important were the histologic parameters tumor type (P = 0.03), tumor grade (P = 0.003), and disease stage (P < 0.0001). In multivariate analysis, disease stage was the most important independent prognostic factor (P < 0.0001), followed by VEGF/KDR (P < 0.01), and VEGF (P < 0.04). Furthermore, VEGF and VEGF/KDR were the only independent prognostic variables for patients with Stage I endometrioid adenocarcinoma. CONCLUSIONS: sMVD and the angiogenic factor VEGF are important indicators of a poor prognosis in patients with endometrial carcinoma. VEGF/KDR complexes define a subgroup of patients with endometrial carcinoma with an even worse prognosis.


Subject(s)
Biomarkers, Tumor/analysis , Endometrial Neoplasms/pathology , Endothelial Growth Factors/biosynthesis , Gene Expression Regulation, Neoplastic , Lymphokines/biosynthesis , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Microcirculation , Middle Aged , Prognosis , Receptors, Vascular Endothelial Growth Factor , Risk Assessment , Survival Analysis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
J Control Release ; 74(1-3): 173-81, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11489493

ABSTRACT

Vascular endothelial growth factor (VEGF) is a primary stimulant of the vascularization of solid tumors and has therefore been the focus of intense research aimed at blocking its activity in solid tumors. VEGF production by tumor cells is induced by oncogenic gene mutations and hypoxic conditions inside the tumor mass. VEGF receptor expression on endothelial cells lining blood vessels in the tumor is also induced by hypoxia and the increased local concentration of VEGF. Therefore in the tumor microenvironment there is an upregulation of both VEGF and its receptor leading to a high concentration of occupied receptor on tumor vascular endothelium. The VEGF-VEGF receptor complex (VEGF-VEGFR) presents an attractive target for the specific delivery of drugs or other effectors to tumor endothelium. Herein we review the development of monoclonal antibodies that selectively bind to the VEGF-VEGFR and their use as targeting agents that selectively bind to VEGF activated blood vessels. Additionally, we summarize the properties of 2C3, a novel monoclonal anti-VEGF antibody that blocks VEGF from binding to VEGFR2 but not VEGFR1. 2C3 may be utilized as both an anti-angiogenic agent by inhibiting VEGFR2 activity and potentially as a vascular targeting agent by binding to blood vessels that express the VEGF-VEGFR1 complex.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Lymphokines/metabolism , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Animals , Biomarkers, Tumor , Endothelial Growth Factors/immunology , Humans , Lymphokines/immunology , Mice , Neoplasms/blood supply , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Growth Factor/immunology , Receptors, Vascular Endothelial Growth Factor , Regional Blood Flow , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
J Pathol ; 194(1): 101-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11329148

ABSTRACT

Active angiogenesis, together with an up-regulation of angiogenic factors, is evident in the synovium of both rheumatoid arthritis (RA) and osteoarthritis (OA). The present study assessed, by immunohistochemistry, the microvessel density in the synovium of these arthritides and in normal controls, in relation to the expression of the angiogenic factors vascular endothelial growth factor (VEGF) and platelet-derived endothelial cell growth factor (PD-ECGF) and the apoptosis-related proteins bcl-2 and p53. More importantly, using the novel 11B5 MAb, the activated "VEGF/flk-1(KDR)-receptor" microvessel density was assessed. VEGF expression in fibroblasts was diffuse in both RA and OA. Diffuse PD-ECGF expression of fibroblasts was noted in all cases of RA, while fibroblast reactivity was focal in the OA material. The standard microvessel density (sMVD), as assessed with the anti-CD31 monoclonal antibody (MAb), was higher in RA (64+/-12) and in OA (65+/-16) than in normal tissues (52+/-8; p=0.008 and 0.0004, respectively). The activated microvessel density (aMVD), assessed with the 11B5 MAb, was significantly higher in RA (29+/-10) than in OA (17+/-4; p<0.0001) and than in normal tissues (14+/-2; p<0.0001). The "activation ratio" (aMVD/sMVD) was statistically higher in RA (0.46+/-0.17) than in OA and normal synovial tissues, the latter two having a similar ratio (0.28+/-0.08 and 0.26+/-0.03, respectively). Cytoplasmic bcl-2 expression was frequent in the synovial cells of OA, but rare in RA. Nuclear p53 protein accumulation was never observed. It is suggested that the angiogenic pathway VEGF/flk-1(KDR) may play an important role in the pathogenesis of RA and OA. Thus, failure of VEGF/flk-1(KDR) activation, in the presence of increased VEGF expression, may indicate a synovium with an impaired capacity to establish a viable vasculature, consistent with the degenerative nature of OA. On the other hand, the activated angiogenesis in RA shows a functional, still pathologically up-regulated VEGF/flk-1(KDR) pathway. Whether restoration of an impaired VEGF/flk-1(KDR) pathway in OA, or inhibition of this in RA, would prove of therapeutic importance requires further investigation.


Subject(s)
Arthritis, Rheumatoid/metabolism , Neovascularization, Pathologic/metabolism , Osteoarthritis/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Synovial Membrane/blood supply , Adult , Aged , Arthritis, Rheumatoid/pathology , Endothelial Growth Factors/metabolism , Female , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , Lymphokines/metabolism , Male , Microcirculation/pathology , Middle Aged , Osteoarthritis/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Mitogen/metabolism , Receptors, Vascular Endothelial Growth Factor , Synovial Membrane/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Biotechniques ; 30(1): 190-4, 196, 198 passim, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11196310

ABSTRACT

Vascular targeting agents (VTAs) can be produced by linking antibodies or antibody fragments directed against endothelial cell markers to effector moieties. So far, it has been necessary to produce the components of VTAs (antibody, antibody fragment, linker, and effector) separately and, subsequently, to conjugate them by biochemical reactions. We devised a cloning and expression system to allow rapid generation of recombinant VTAs from hybridoma cell lines. The VTAs consist of a single chain Fv antibody fragment as a targeting moiety and either truncated Pseudomonas exotoxin (resulting in immunotoxins) or truncated human tissue factor (resulting in coaguligands) as effectors. The system was applied to generate recombinant immunotoxins and coaguligands directed against endoglin, vascular endothelial growth factor (VEGF):VEGF receptor (VEGFR) complex and vascular cell adhesion molecule 1 (VCAM-1). The fusion proteins exhibited similar functional activity to analogous biochemical constructs. This is the first report to describe the generation and characterization of recombinant coaguligands.


Subject(s)
Immunoglobulin Fragments/pharmacology , Immunotoxins/pharmacology , Neovascularization, Pathologic/prevention & control , Thromboplastin/pharmacology , Animals , Antigens, CD , Cell Division/drug effects , Cell Line , Cloning, Molecular , Dose-Response Relationship, Drug , Endoglin , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endothelial Growth Factors/immunology , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Factor Xa/drug effects , Factor Xa/metabolism , Female , Humans , Hybridomas , Immunoglobulin Fragments/genetics , Immunohistochemistry , Immunotoxins/genetics , Lymphokines/immunology , Lymphokines/metabolism , Mice , Plasmids/genetics , Protein Binding , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface , Receptors, Growth Factor/immunology , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Thromboplastin/genetics , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Anticancer Res ; 21(6B): 4221-9, 2001.
Article in English | MEDLINE | ID: mdl-11908675

ABSTRACT

Vascular targeting agents, which selectively destroy tumor blood vessels, are attractive agents for the treatment of solid tumors. They differ from anti-angiogenic agents in that they target the mature, blood-conducting vessels of the tumors. They are better suited for larger tumors where angiogenesis can occur less frequently. For application in man, target molecules are needed that are selectively expressed on the vascular endothelium of tumors. Such markers include the complexes that are formed when vascular endothelial growth factor (VEGF) binds to its receptors (VEGFR). VEGF production by tumor cells is induced by oncogenic gene mutations and by the hypoxic conditions within the tumor mass. The receptors, VEGFR1 (FLT-1) and VEGFR2 (KDR/Flk-1), are upregulated on vascular endothelial cells in tumors by hypoxia and by the increased local concentration of VEGF. Consequently, there is a high concentration of occupied receptors on tumor vascular endothelium. Here, we review the concept of vascular targeting and the development of monoclonal antibodies that bind to VEGF: VEGFR complexes and their use as tumor vascular targeting agents. A promising monoclonal antibody is 2C3, which blocks VEGF from binding to VEGFR2 but not VEGFR1. We conclude that 2C3 might have dual activity as an anti-angiogenic agent by inhibiting VEGFR2 activity and as a vascular targeting agent for selective drug delivery to tumor vessels.


Subject(s)
Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Endothelial Growth Factors/immunology , Endothelial Growth Factors/metabolism , Humans , Lymphokines/immunology , Lymphokines/metabolism , Neovascularization, Pathologic/immunology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Cancer Res ; 60(18): 5117-24, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016638

ABSTRACT

Vascular endothelial growth factor (VEGF) is a multifunctional angiogenic growth factor that is a primary stimulant of the development and maintenance of a vascular network in embryogenesis and the vascularization of solid tumors. At the present time there are two well-characterized receptors for VEGF that are selectively expressed on endothelium. VEGF receptor 2 [VEGFR2 (KDR/Flk-1)] mediates endothelial cell mitogenesis and permeability increases, whereas the role of VEGF receptor 1 [VEGFR1 (Flt-1)] has not been clearly defined. In the present study, a monoclonal antibody, 2C3, is shown to block the interaction of VEGF with VEGFR2 but not with VEGFR1 through ELISA, receptor binding assays, and receptor activation assays. 2C3 blocks the VEGF-induced vascular permeability increase in guinea pig skin. 2C3 has potent antitumor activity, inhibiting the growth of newly injected and established human tumor xenografts in mice. These findings demonstrate the usefulness of 2C3 in dissecting the pathways that are activated by VEGF in cells that express both VEGFR1 and VEGFR2, as well as highlighting the dominant role of VEGFR2 in mediating VEGF-induced vascular permeability increase and tumor angiogenesis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Endothelial Growth Factors/immunology , Growth Inhibitors/pharmacology , Lymphokines/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Capillary Permeability/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Division/drug effects , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Fibrosarcoma/pathology , Fibrosarcoma/therapy , Growth Inhibitors/immunology , Guinea Pigs , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Lymphokines/antagonists & inhibitors , Lymphokines/metabolism , Male , Mice , Neoplasm Transplantation , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/therapy , Skin/blood supply , Substrate Specificity , Transplantation, Heterologous , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
17.
Cancer Res ; 60(11): 3088-95, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10850461

ABSTRACT

Vascular endothelial growth factor (VEGF) is an important angiogenic factor, linked to poor outcome in human malignancies including non-small cell lung carcinoma (NSCLC). We used the 11B5 monoclonal antibody recognizing the VEGF/KDR complex (R. A. Brekken et al., Cancer Res., 58: 1952-1959, 1998) to assess the VEGF expression in cancer cells and the VEGF/KDR activated microvessel density (aMVD) in early operable NSCLC. The JC70 anti-CD31 monoclonal antibody was used to assess the standard MVD (sMVD). The aMVD was significantly higher in the invading front of the tumors and in the normal lung adjacent to the tumors as compared with normal lung distant to the tumor or to inner tumor areas (P < 0.0002). The sMVD was higher in the normal lung and decreased from the invading front to inner tumor areas (P < 0.0001). However, the vascular activation (aMVD:sMVD) was 4-6 times higher in the tumor areas as compared with lung from normal individuals (36-58% versus 9%; P < 0.0001). Fibroblast 11B5 reactivity, noted in 25% of cases, correlated with high aMVD and sMVD in the inner tumor areas. Multivariate analysis showed that aMVD was the most potent and independent prognostic factor (P = 0.001; t-ratio, 3.28). It is concluded that intense VEGF/KDR angiogenic pathway activation is a tumor-specific feature in more than 50% of NSCLC cases and is associated with poor postoperative outcome. Clinical trials involving targeting of the VEGF/KDR-positive vasculature with specific antibodies, such as 11B5, are, therefore, encouraged.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Endothelial Growth Factors/biosynthesis , Lung Neoplasms/metabolism , Lymphokines/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Adenocarcinoma/metabolism , Adult , Aged , Antibodies, Monoclonal/metabolism , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/metabolism , Female , Fibroblasts/metabolism , Humans , Immunohistochemistry , Lung/metabolism , Lung Neoplasms/blood supply , Lung Neoplasms/mortality , Male , Microcirculation , Middle Aged , Necrosis , Prognosis , Receptors, Vascular Endothelial Growth Factor , Time Factors , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Clin Cancer Res ; 6(5): 1917-21, 2000 May.
Article in English | MEDLINE | ID: mdl-10815916

ABSTRACT

We investigated the expression of MUC1 protein and its relationship to the microvessel density and the expression of thymidine phosphorylase, vascular endothelial growth factor (VEGF), VEGF-receptor KDR, basic fibroblast growth factor (bFGF), and bFGF-receptor (FGFR-2) in non-small cell lung cancer. Although MUC1 expression was found equally in poorly and highly vascularized tumors, a significant coexpression with multiple angiogenic factors and their receptors was noted (P = 0.0002, 0.03, 0.19, 0.10, and 0.01 for thymidine phosphorylase, VEGF, KDR, bFGF, and FGFR-2, respectively). In multiple regression analysis, both angiogenesis and MUC1 expression were independent prognostic variables. The present study suggests the existence of an early genetic event leading to the activation of both migration and angiogenesis pathways in non-small cell lung cancer.


Subject(s)
Angiogenesis Inducing Agents/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Mucin-1/biosynthesis , Adult , Aged , Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/pathology , Endothelial Growth Factors/biosynthesis , Female , Fibroblast Growth Factor 2/biosynthesis , Humans , Immunohistochemistry , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Lymphokines/biosynthesis , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neovascularization, Pathologic , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/biosynthesis , Receptors, Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Thymidine Phosphorylase/biosynthesis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
20.
J Histochem Cytochem ; 48(4): 545-56, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727296

ABSTRACT

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) interacts with two high-affinity tyrosine kinase receptors, VEGFR-1 and VEGFR-2, to increase microvascular permeability and induce angiogenesis. Both receptors are selectively expressed by vascular endothelial cells and are strikingly increased in tumor vessels. We used a specific antibody to localize VEGFR-2 (FLK-1, KDR) in microvascular endothelium of normal mouse kidneys and in the microvessels induced by the TA3/St mammary tumor or by infection with an adenoviral vector engineered to express VPF/VEGF. A pre-embedding method was employed at the light and electron microscopic levels using either nanogold or peroxidase as reporters. Equivalent staining was observed on both the luminal and abluminal surfaces of tumor- and adenovirus-induced vascular endothelium, but plasma membranes at interendothelial junctions were spared except at sites connected to vesiculovacuolar organelles (VVOs). VEGFR-2 was also localized to the membranes and stomatal diaphragms of some VVOs. This staining distribution is consistent with a model in which VPF/VEGF increases microvascular permeability by opening VVOs to allow the transendothelial cell passage of plasma and plasma proteins.


Subject(s)
Capillary Permeability , Genetic Vectors/metabolism , Kidney/metabolism , Mammary Neoplasms, Experimental/blood supply , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Adenoviridae/genetics , Animals , Capillaries/metabolism , Endothelium, Vascular/metabolism , Female , Kidney/blood supply , Kidney/ultrastructure , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Kidney Glomerulus/ultrastructure , Kidney Tubules/blood supply , Kidney Tubules/metabolism , Kidney Tubules/ultrastructure , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Microscopy, Electron , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...