Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Curr Neurovasc Res ; 16(3): 258-265, 2019.
Article in English | MEDLINE | ID: mdl-31258081

ABSTRACT

OBJECTIVE: This study aimed to examine whether DC101 (anti-VEGFR2 antibody)- modified micelles have applications as novel drug delivery devices, which allow small molecule antiangiogenic agents to deliver to angiogenic sites on a murine laser-induced choroidal neovascularization (CNV) model. MATERIALS AND METHODS: CNV was induced by photocoagulation on the unilateral eye of each mouse under anesthesia. Immediately after laser coagulation, E7974-loaded DC101-modified micelles and motesanib-loaded DC101-modified micelles were intravitreally administrated. Two weeks after photocoagulation, CNV was visualized using fluorescein-conjugated dextran (MW=2,000 kDa), and the CNV area was measured in retinal pigment epithelium (RPE)-choroidal flat mounts. RESULTS: Intravitreal administration of both DC101-modified micelles loaded with E7974 at 2 µM and motesanib at 2 µM significantly reduced CNV area in the murine laser-induced CNV model at a clearly lower concentration than the effective dose of each agent. CONCLUSION: These results suggest that DC101-modified micelle might be effective drug carrier system for treating CNV and other ocular angiogenic diseases.


Subject(s)
Autoantibodies/administration & dosage , Choroidal Neovascularization/drug therapy , Drug Delivery Systems/methods , Micelles , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Animals , Autoantibodies/metabolism , Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Intravitreal Injections/methods , Lasers/adverse effects , Male , Mice , Mice, Inbred C57BL , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
2.
Clin Exp Pharmacol Physiol ; 44(1): 71-78, 2017 01.
Article in English | MEDLINE | ID: mdl-27562635

ABSTRACT

In order to achieve a synergistic effect on anti-tumour and anti-angiogenesis activity, we designed and constructed a DNA vaccine that expresses MUC1and VEGFR2 in the same reading frame. The aim of this study was to investigate the anti-tumour activity of this DNA vaccine. Furthermore, we also investigated the enhanced synergistic anti-Lewis lung carcinoma effect of this DNA vaccine by using GM-CSF as an adjuvant. A series of DNA plasmids encoding MUC1, VEGFR2, GM-CSF, and their conjugates were constructed and injected into mice intramuscularly (i.m.) followed by an electric pulse. The humoral and cellular immune responses after immunization were detected by enzyme-linked immunosorbent assay (ELISA) and enzyme-linked immunospot (ELISPOT), respectively. To evaluate the anti-tumour efficacy of these plasmids, murine models with MUC1-expressing tumours were generated. After injection into the tumour-bearing mouse model, the plasmid carrying the fusion gene of MUC1 and VEGFR2 showed stronger inhibition of tumour growth than the plasmid expressing MUC1 or VEGFR2 alone, which indicated that MUC1 and VEGFR2 could exert a synergistic anti-tumour effect. Furthermore, mice vaccinated with the combination of the GM-CSF expressing plasmid and the plasmid carrying the fusion gene of MUC1 and VEGFR2 showed an increased inhibition in the growth of MUC1-expressing tumours and prolonged mouse survival. These observations emphasize the potential of the synergistic anti-tumour and anti-angiogenesis strategy used in DNA vaccines, and the potential of the GM-CSF gene as an adjuvant for DNA vaccines, which could represent a promising approach for tumour immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Mucin-1/administration & dosage , Vaccines, DNA/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Animals , Carcinoma, Lewis Lung/immunology , Cells, Cultured , Chemotherapy, Adjuvant/methods , Drug Synergism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Mice , Mice, Inbred C57BL , Mucin-1/genetics , Mucin-1/immunology , Spleen/drug effects , Spleen/immunology , Treatment Outcome , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology
3.
Molecules ; 21(5)2016 Apr 29.
Article in English | MEDLINE | ID: mdl-27136525

ABSTRACT

In this study, we report on the design, synthesis, photokinetic properties and in vitro evaluation of photoactivatable caged prodrugs for the receptor tyrosine kinase VEGFR-2. Highly potent VEGFR-2 inhibitors 1 and 3 were caged by introduction of a photoremovable protecting group (PPG) to yield the caged prodrugs 4 and 5. As expected, enzymatic and cellular proliferation assays showed dramatically diminished efficacy of caged prodrugs in vitro. Upon ultraviolet (UV) irradiation of the prodrugs original inhibitory activity was completely restored and even distinctly reinforced, as was the case for the prodrug 4. The presented results are a further evidence for caging technique being an interesting approach in the protein kinase field. It could enable spatial and temporal control for the inhibition of VEGFR-2. The described photoactivatable prodrugs might be highly useful as biological probes for studying the VEGFR-2 signal transduction.


Subject(s)
Prodrugs , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Cell Line , Cell Proliferation/drug effects , Drug Design , Molecular Structure , Photochemical Processes , Prodrugs/radiation effects , Ultraviolet Rays , Vascular Endothelial Growth Factor Receptor-2/administration & dosage
4.
Cell Death Dis ; 7: e2212, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27148685

ABSTRACT

There is increasing evidence that VEGF-A antagonists may be detrimental to neuronal health following ocular administration. Here we investigated firstly the effects of VEGF-A neutralization on retinal neuronal survival in the Ins2(Akita) diabetic and JR5558 spontaneous choroidal neovascularization (CNV) mice, and then looked at potential mechanisms contributing to cell death. We detected elevated apoptosis in the ganglion cell layer in both these models following VEGF-A antagonism, indicating that even when vascular pathologies respond to treatment, neurons are still vulnerable to reduced VEGF-A levels. We observed that retinal ganglion cells (RGCs) seemed to be the cells most susceptible to VEGF-A antagonism, so we looked at anterograde transport in these cells, due to their long axons requiring optimal protein and organelle trafficking. Using cholera toxin B-subunit tracer studies, we found a distal reduction in transport in the superior colliculus following VEGF-A neutralization, which occurred prior to net RGC loss. This phenomenon of distal transport loss has been described as a feature of early pathological changes in glaucoma, Alzheimer's and Parkinson's disease models. Furthermore, we observed increased phosphorylation of p38 MAPK and downstream Hsp27 stress pathway signaling in the retinas from these experiments, potentially providing a mechanistic explanation for our findings. These experiments further highlight the possible risks of using VEGF-A antagonists to treat ocular neovascular disease, and suggest that VEGF-A may contribute to the maintenance and function of axonal transport in neurons of the retina.


Subject(s)
Choroidal Neovascularization/genetics , Diabetes Mellitus, Experimental/genetics , Retinal Ganglion Cells/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/adverse effects , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Axonal Transport/drug effects , Cell Tracking , Cholera Toxin/chemistry , Cholera Toxin/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Gene Expression Regulation , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Intravitreal Injections , Male , Mice , Mice, Transgenic , Phosphorylation/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Cancer Sci ; 106(7): 883-90, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25867139

ABSTRACT

Gemcitabine is a key drug for the treatment of pancreatic cancer; however, with its limitation in clinical benefits, the development of another potent therapeutic is necessary. Vascular endothelial growth factor receptor 2 is an essential target for tumor angiogenesis, and we have conducted a phase I clinical trial using gemcitabine and vascular endothelial growth factor receptor 2 peptide (elpamotide). Based on the promising results of this phase I trial, a multicenter, randomized, placebo-controlled, double-blind phase II/III clinical trial has been carried out for pancreatic cancer. The eligibility criteria included locally advanced or metastatic pancreatic cancer. Patients were assigned to either the Active group (elpamotide + gemcitabine) or Placebo group (placebo + gemcitabine) in a 2:1 ratio by the dynamic allocation method. The primary endpoint was overall survival. The Harrington-Fleming test was applied to the statistical analysis in this study to evaluate the time-lagged effect of immunotherapy appropriately. A total of 153 patients (Active group, n = 100; Placebo group, n = 53) were included in the analysis. No statistically significant differences were found between the two groups in the prolongation of overall survival (Harrington-Fleming P-value, 0.918; log-rank P-value, 0.897; hazard ratio, 0.87, 95% confidence interval [CI], 0.486-1.557). Median survival time was 8.36 months (95% CI, 7.46-10.18) for the Active group and 8.54 months (95% CI, 7.33-10.84) for the Placebo group. The toxicity observed in both groups was manageable. Combination therapy of elpamotide with gemcitabine was well tolerated. Despite the lack of benefit in overall survival, subgroup analysis suggested that the patients who experienced severe injection site reaction, such as ulceration and erosion, might have better survival.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Adenosquamous/drug therapy , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Cancer Vaccines/administration & dosage , Carcinoma, Adenosquamous/mortality , Carcinoma, Adenosquamous/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Double-Blind Method , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Peptide Fragments/administration & dosage , Treatment Outcome , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Gemcitabine
6.
Biomacromolecules ; 15(3): 756-62, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24460101

ABSTRACT

Heterogeneous toroidal-spiral particles (TSPs) were generated by polymer droplet sedimentation, interaction, and cross-linking. TSPs provide a platform for encapsulation and release of multiple compounds of different sizes and physicochemical properties. As a model system, we demonstrate the encapsulation and independently controlled release of an anti-VEGFR-2 antibody and irinotecan for the treatment of glioblastoma multiforme. The anti-VEGFR-2 antibody was released from the TS channels and its binding to HUVECs was confirmed by confocal microscopy and flow cytometry, suggesting active antibody encapsulation and release. Irinotecan, a small molecule drug, was released from the dense polymer matrix of poly(ethylene glycol) diacrylate (MW ~ 700 g/mol; PEGDA 700). Released irinotecan inhibited the proliferation of U251 malignant glioma cells. Since the therapeutic compounds are released through different pathways, specifically diffusion through the polymer matrix versus TS channels, the release rate can be controlled independently through the design of the structure and material of particle components.


Subject(s)
Antibodies, Anti-Idiotypic/administration & dosage , Drug Delivery Systems , Glioblastoma/drug therapy , Vascular Endothelial Growth Factor Receptor-2/immunology , Antibodies, Anti-Idiotypic/chemistry , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Glioblastoma/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Irinotecan , Particle Size , Polymers/chemistry , Vascular Endothelial Growth Factor Receptor-2/administration & dosage
7.
Mol Cancer Ther ; 12(10): 2055-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23858102

ABSTRACT

The serine protease granzyme B (GrB) induces apoptosis through both caspase-dependent and -independent multiple-cascade mechanisms. VEGF121 binds to both VEGF receptor (VEGFR)-1 and VEGFR-2 receptors. We engineered a unique GrB/VEGF121 fusion protein and characterized its properties in vitro and in vivo. Endothelial and tumor cell lines showed varying levels of sensitivity to GrB/VEGF121 that correlated closely to total VEGFR-2 expression. GrB/VEGF121 localized efficiently into VEGFR-2-expressing cells, whereas the internalization into VEGFR-1-expressing cells was significantly reduced. Treatment of VEGFR-2(+) cells caused mitochondrial depolarization in 48% of cells by 48 hours. Exposure to GrB/VEGF121 induced apoptosis in VEGFR-2(+), but not in VEGFR-1(+), cells and rapid caspase activation was observed that could not be inhibited by treatment with a pan-caspase inhibitor. In vivo, GrB/VEGF121 localized in perivascular tumor areas adjacent to microvessels and in other areas in the tumor less well vascularized, whereas free GrB did not specifically localize to tumor tissue. Administration (intravenous) of GrB/VEGF121 to mice at doses up to 40 mg/kg showed no toxicity. Treatment of mice bearing established PC-3 tumor xenografts with GrB/VEGF121 showed significant antitumor effect versus treatment with GrB or saline. Treatment with GrB/VEGF121 at 27 mg/kg resulted in the regression of four of five tumors in this group. Tumors showed a two-fold lower Ki-67-labeling index compared with controls. Our results show that targeted delivery of GrB to tumor vascular endothelial cells or to tumor cells activates apoptotic cascades and this completely human construct may have significant therapeutic potential.


Subject(s)
Granzymes/genetics , Neoplasms/drug therapy , Recombinant Fusion Proteins/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Administration, Intravenous , Animals , Apoptosis/drug effects , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Granzymes/administration & dosage , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Recombinant Fusion Proteins/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/administration & dosage
8.
Oncol Rep ; 29(4): 1510-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404510

ABSTRACT

As tumor-associated antigens are not well characterized for the majority of human tumors, polyvalent vaccines prepared with whole-tumor antigens are an attractive approach for tumor vaccination. Vascular endothelial growth factor receptor-2 (VEGFR2), as a model antigen with which to explore the feasibility of immunotherapy, has shown great promise as a tumor vaccine. However, the efficacy of immunotherapy is often not ideal when used alone. In this study, we explored the therapeutic efficacy of an irradiated AdVEGFR2-infected cell vaccine-based immunotherapy in the weakly immunogenic and highly metastatic 4T1 murine mammary cancer model. An adenovirus encoding the VEGFR2 gene (AdVEGFR2) was constructed. Lethally irradiated, virus-infected 4T1 cells were used as vaccines. Vaccination with lethally irradiated AdVEGFR2-infected 4T1 cells inhibited subsequent tumor growth and pulmonary metastasis compared with challenge inoculations. Angiogenesis was inhibited, and the number of CD8+ T lymphocytes was increased within the tumors. Antitumor activity was also caused by the adoptive transfer of isolated spleen lymphocytes. In vitro, the expression of HMGB1 and HSP70 in the AdVEGFR2­infected 4T1 cells was increased, and was involved in the activation of tumor antigen-specific T-cell immunity. Our results indicate that the immunotherapy based on irradiated AdVEGFR2-infected whole-cancer cell vaccines may be a potentially effective strategy for 4T1 cancer treatment.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Fibroadenoma/therapy , Immunotherapy, Active , Vascular Endothelial Growth Factor Receptor-2/genetics , Adenoviridae/genetics , Adenoviridae/radiation effects , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Female , Fibroadenoma/immunology , Fibroadenoma/pathology , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neovascularization, Pathologic/immunology , Vascular Endothelial Growth Factor Receptor-2/administration & dosage
9.
Cancer Sci ; 103(12): 2135-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22957712

ABSTRACT

Targeting of tumor angiogenesis with vaccines is a potentially valuable approach to cancer treatment. Elpamotide is an immunogenic peptide derived from vascular endothelial growth factor receptor 2, which is expressed at a high level in vascular endothelial cells. We have now carried out a phase I study to evaluate safety, the maximum tolerated dose, and potential pharmacodynamic biomarkers for this vaccine. Ten HLA-A*24:02-positive patients with advanced refractory solid tumors received elpamotide s.c. at dose levels of 0.5, 1.0, or 2.0 mg once a week on a 28-day cycle. Five patients experienced an injection site reaction of grade 1 and 2, which was the most frequent adverse event. In the 1.0 mg cohort, one patient experienced proteinuria of grade 1 and another patient developed both hypertension and proteinuria of grade 1. No adverse events of grade 3 or higher were observed, and the maximum tolerated dose was therefore not achieved. The serum concentration of soluble vascular endothelial growth factor receptor 2 decreased significantly after elpamotide vaccination. Microarray analysis of gene expression in PBMCs indicated that several pathways related to T cell function and angiogenesis were affected by elpamotide vaccination, supporting the notion that this peptide induces an immune response that targets angiogenesis in the clinical setting. In conclusion, elpamotide is well tolerated and our biomarker analysis indicates that this anti-angiogenic vaccine is biologically active. Clinical trial registration no. UMIN000008336.


Subject(s)
Cancer Vaccines/therapeutic use , Peptide Fragments/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/therapeutic use , Aged , Cancer Vaccines/administration & dosage , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , HLA-A24 Antigen/metabolism , Humans , Male , Maximum Tolerated Dose , Middle Aged , Peptide Fragments/administration & dosage , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/administration & dosage
10.
Vaccine ; 29(34): 5802-11, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21482223

ABSTRACT

Vascular endothelial growth factor (VEGF) has been known as a potential vasculogenic and angiogenic factor and its receptor (VEGFR2) is a major receptor to response to the angiogenic activity of VEGF. The technique that to break the immune tolerance of "self-antigens" associated with angiogenesis is an attractive approach for cancer therapy with T4 phage display system. In this experiment, mouse VEGFR2 was constructed on T4 phage nanometer-particle surface as a recombinant vaccine. T4-mVEGFR2 recombinant vaccine was identified by PCR and western blot assay. Immunotherapy with T4-mVEGFR2 was confirmed by protective immunity against Lewis lung carcinoma (LLC) in mice. The antibody against mVEGFR2 was detected by ELISPOT, ELISA and Dot ELISA. The inhibitive effects against angiogenesis were studied using CD31 and CD105 via histological analysis. VEGF-mediated endothelial cells proliferation and tube formation were inhibited in vitro by immunoglobulin induced by T4-mVEGFR2. The antitumor activity was substantiated from the adoptive transfer of the purified immunoglobulin. Antitumor activity and autoantibody production of mVEGFR2 could be neutralized by the depletion of CD4+T lymphocytes. These studies strongly suggest that T4-mVEGFR2 recombinant vaccine might be a promising antitumor approach.


Subject(s)
Carcinoma, Lewis Lung , Neovascularization, Pathologic , Vaccines, Synthetic , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/immunology , Adoptive Transfer , Angiogenesis Inhibitors/administration & dosage , Animals , Bacteriophage T4/genetics , Bacteriophage T4/immunology , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Cell Proliferation , Endoglin , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Immunotherapy/methods , Intracellular Signaling Peptides and Proteins/blood , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Neovascularization, Pathologic/therapy , Platelet Endothelial Cell Adhesion Molecule-1/blood , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/pharmacology
11.
Vaccine ; 28(44): 7221-7, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20800113

ABSTRACT

Although the critical role of complement component C3d as a molecular adjuvant in preventing virus infection is well established, its role in cancer therapies is unclear. In this study, we have engineered a DNA vaccine that expresses extracellular region of murine VEGFR-2 (FLK1(265-2493)) and 3 copies of C3d (C3d3), a component of complement as a molecular adjuvant, designed to increase antitumor immunity. VEGFR-2 has a more restricted expression on endothelial cells and is upregulated once these cells proliferate during angiogenesis in the tumor vasculature. Immunization of mice with vector encoding FLK1(265-2493) alone generated only background levels of anti-VEGFR-2 antibodies and slight inhibitory effect on tumor growth. However, the addition of C3d3 to the vaccine construct significantly augmented the anti-VEGFR-2 humoral immune response and inhibited the tumor growth. The antitumor activity induced by vaccination with vector encoding FLK1(265-2493)-C3d3 fusion protein was also demonstrated via growth inhibition of established tumors following passive transfer of immune serum from vaccinated mice. Our results suggest that vaccination with vector encoding FLK1(265-2493) with C3d3 as a molecular adjuvant induces adaptive humoral activity, which is directed against the murine VEGFR-2 and can significantly inhibit tumor growth, and that administration of C3d as a molecular adjuvant to increase antibodies levels to VEGFR-2 may provide an alternative treatment modality for cancer therapies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Complement C3d/administration & dosage , Melanoma, Experimental/therapy , Vaccines, DNA/immunology , Animals , Complement C3d/immunology , Endothelial Cells/immunology , Female , HeLa Cells , Humans , Immunity, Humoral , Immunotherapy, Adoptive , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/immunology
12.
Clin Cancer Res ; 16(2): 664-72, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068097

ABSTRACT

PURPOSE: To evaluate clinical activity and target modulation of vandetanib in women with recurrent ovarian cancer. EXPERIMENTAL DESIGN: A phase II trial of orally administered vandetanib 300 mg daily was designed to include analyses of target inhibition through paired biopsies and dynamic imaging. Core 18-gauge needle biopsies and dynamic contrast-enhanced magnetic resonance imaging were obtained before initiation of therapy and 6 weeks into therapy. Biopsy samples were subjected to reverse-phase protein lysate array endpoint analysis. Cytokine concentrations were measured by enzyme-linked immunosorbent assay in serially collected plasma samples. RESULTS: Twelve patients entered the study, and accrual was terminated in the first stage because of lack of response or disease stabilization beyond 6 months. Adverse events included rash, diarrhea, and prolonged QT interval corrected for heart rate, but not hypertension. Exploratory analyses showed that epidermal growth factor receptor (EGFR) phosphorylation was reduced in the eight paired biopsy sets obtained; vascular endothelial growth factor (VEGF) receptor-2 phosphorylation was not consistently affected nor were dynamic contrast-enhanced MRI permeability and flow parameters. Serial plasma VEGF concentrations were variable and did not significantly change in the 11 patients assessed. CONCLUSIONS: Vandetanib 300 mg daily monotherapy had no significant clinical benefit in this disease setting. Proteomic analysis of paired biopsies detected both phosphorylated-EGFR and phosphorylated-VEGF receptor-2 in ovarian tumor tissue, but only phosphorylated-EGFR was measurably inhibited by vandetanib.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , ErbB Receptors/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Piperidines/therapeutic use , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Drug Administration Schedule , ErbB Receptors/metabolism , Female , Humans , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Piperidines/administration & dosage , Piperidines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/administration & dosage , Quinazolines/pharmacology , Recurrence , Signal Transduction/drug effects , Treatment Failure , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
J Immunol ; 182(9): 5537-46, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380802

ABSTRACT

Thirty years after angiogenesis was shown to play an enabling role in cancer, modern medicine is still trying to develop novel compounds and therapeutics to target the tumor vasculature. However, most therapeutics require multiple rounds of administration and can have toxic side effects. In this study, we use anti-angiogenesis immunotherapy to target cells actively involved in forming new blood vessels that support the growth and spread of breast cancer. Targeting a central cell type involved in angiogenesis, endothelial cells, we immunized against host vascular endothelial growth factor receptor 2 to fight the growth of Her-2/neu(+) breast tumors. Using the bacterial vector, Listeria monocytogenes (Lm), we fused polypeptides from the mouse vascular endothelial growth factor receptor 2 molecule (fetal liver kinase-1) to the microbial adjuvant, listeriolysin-O, and used Lm to deliver the Ags and elicit potent antitumor CTL responses. Lm-listeriolysin-O-fetal liver kinase-1 was able to eradicate some established breast tumors, reduce microvascular density in the remaining tumors, protect against tumor rechallenge and experimental metastases, and induce epitope spreading to various regions of the tumor-associated Ag Her-2/neu. Tumor eradication was found to be dependent on epitope spreading to HER-2/neu and was not solely due to the reduction of tumor vasculature. However, vaccine efficacy did not affect normal wound healing nor have toxic side effects on pregnancy. We show that an anti-angiogenesis vaccine can overcome tolerance to the host vasculature driving epitope spreading to an endogenous tumor protein and drive active tumor regression.


Subject(s)
Angiogenesis Inhibitors/immunology , Cancer Vaccines/immunology , Listeria monocytogenes/immunology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/prevention & control , Neovascularization, Pathologic/prevention & control , Receptor, ErbB-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/immunology , Amino Acid Sequence , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/genetics , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Female , Growth Inhibitors/administration & dosage , Growth Inhibitors/genetics , Growth Inhibitors/immunology , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Listeria monocytogenes/genetics , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/physiopathology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/genetics
14.
J Neurosurg ; 108(5): 979-88, 2008 May.
Article in English | MEDLINE | ID: mdl-18447716

ABSTRACT

OBJECT: Glioblastoma multiforme (GBM) is characterized by neovascularization, raising the question of whether angiogenic blockade may be a useful therapeutic strategy for this disease. It has been suggested, however, that, to be useful, angiogenic blockade must be persistent and at levels sufficient to overcome proangiogenic signals from tumor cells. In this report, the authors tested the hypothesis that sustained high concentrations of 2 different antiangiogenic proteins, delivered using a systemic gene therapy strategy, could inhibit the growth of established intracranial U87 human GBM xenografts in nude mice. METHODS: Mice harboring established U87 intracranial tumors received intravenous injections of adenoviral vectors encoding either the extracellular domain of vascular endothelial growth factor receptor-2-Fc fusion protein (Ad-VEGFR2-Fc) alone, soluble endostatin (Ad-ES) alone, a combination of Ad-VEGFR2-Fc and Ad-ES, or immunoglobulin 1-Fc (Ad-Fc) as a control. RESULTS: Three weeks after treatment, magnetic resonance imaging-based determination of tumor volume showed that treatment with Ad-VEGFR2-Fc, Ad-ES, or Ad-VEGFR2-Fc in combination with Ad-ES, produced 69, 59, and 74% growth inhibition, respectively. Bioluminescent monitoring of tumor growth revealed growth inhibition in the same treatment groups to be 62, 74, and 72%, respectively. Staining with proliferating cell nuclear antigen and with terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling showed reduced tumor cell proliferation and increased apoptosis in all antiangiogenic treatment groups. CONCLUSIONS: These results suggest that systemic delivery and sustained production of endostatin and soluble VEGFR2 can slow intracranial glial tumor growth by both reducing cell proliferation and increasing tumor apoptosis. This work adds further support to the concept of using antiangiogenesis therapy for intracranial GBM.


Subject(s)
Brain Neoplasms/pathology , Endostatins/administration & dosage , Glioblastoma/pathology , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Adenoviridae , Animals , Apoptosis , Endostatins/analysis , Endostatins/genetics , Genetic Vectors , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/analysis , Vascular Endothelial Growth Factor Receptor-2/genetics
15.
J Immunol ; 177(6): 4122-31, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16951377

ABSTRACT

Stromal keratitis (SK) is an immunoinflammatory eye lesion caused by HSV-1 infection. One essential step in the pathogenesis is neovascularization of the normally avascular cornea, a process that involves the vascular endothelial growth factor (VEGF) family of proteins. In this report, we targeted the proliferating vascular endothelial cells expressing VEGFR-2 in the SK cornea by immunization with recombinant Salmonella typhimurium containing a plasmid encoding murine VEGFR-2. This form of DNA immunization resulted in diminished angiogenesis and delayed development of SK caused by HSV-1 infection and also reduced angiogenesis resulting from corneal implantation with rVEGF. CTL responses against endothelial cells expressing VEGFR-2 were evident in the VEGFR-2-immunized group and in vivo CD8+ T cell depletion resulted in the marked reduction of the antiangiogenic immune response. These results indicate a role for CD8+ T cells in the antiangiogenic effects. Our results may also imply that the anti-VEGFR-2 vaccination approach might prove useful to control pathological ocular angiogenesis and its consequences.


Subject(s)
Angiogenesis Inhibitors/immunology , Cornea/blood supply , Keratitis, Herpetic/prevention & control , Neovascularization, Pathologic/prevention & control , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/genetics , Animals , Cells, Cultured , Cornea/pathology , Cornea/virology , Female , Genetic Vectors , Herpesvirus 1, Human/immunology , Keratitis, Herpetic/immunology , Keratitis, Herpetic/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/virology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Stromal Cells/immunology , Stromal Cells/pathology , Stromal Cells/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
16.
Ann Surg Oncol ; 12(12): 1025-36, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16244806

ABSTRACT

BACKGROUND: Tumor hypoxia induces vascular endothelial growth factor (VEGF) expression, which stimulates angiogenesis and tumor proliferation. The VEGF signaling pathway is inhibited by soluble VEGF receptors (soluble fetal liver kinase 1; sFlk-1), which bind VEGF and block its interaction with endothelial cells. Herpes simplex virus (HSV) amplicons are replication-incompetent viruses used for gene delivery. We attempted to attenuate angiogenesis and inhibit pancreatic tumor growth through HSV amplicon-mediated expression of sFlk-1 under hypoxic control. METHODS: A multimerized hypoxia-responsive enhancer (10 x HRE) was cloned upstream of the sFlk-1 gene (10 x HRE/sFlk-1). A novel HSV amplicon expressing 10 x HRE/sFlk-1 was genetically engineered (HSV10 x HRE/sFlk-1).Human pancreatic adenocarcinoma cells (AsPC1) were transduced with HSV10 x HRE/sFlk-1 and incubated in normoxia (21% oxygen) or hypoxia (1% oxygen). Capillary inhibition was evaluated by human umbilical vein endothelial cell assay. Western blot assessed sFlk-1 expression. AsPC1 flank tumor xenografts (n = 24) were transduced with HSV10 x HRE/sFlk-1. RESULTS: Media from normoxic AsPC1 transduced with HSV10 x HRE/sFlk-1 yielded a 36% reduction in capillary formation versus controls (P < .05), whereas hypoxic AsPC1 yielded a 76% reduction (P < .005). Western blot of AsPC1 transduced with HSV10 x HRE/sFlk-1 demonstrated greater sFlk-1 expression in hypoxia versus normoxia. AsPC1 flank tumors treated with HSV10 x HRE/sFlk-1 exhibited a 59% reduction in volume versus controls (P < .000001). CONCLUSIONS: HSV amplicon delivery of a hypoxia-inducible soluble VEGF receptor significantly reduces new vessel formation and tumor growth. Tumor hypoxia can thus be used to direct antiangiogenic therapy to pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/therapy , Angiogenesis Inhibitors/administration & dosage , Hypoxia-Inducible Factor 1 , Neovascularization, Physiologic/drug effects , Pancreatic Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Adenocarcinoma/pathology , Blotting, Western , Cell Hypoxia , Enhancer Elements, Genetic , Genetic Vectors/genetics , Humans , Pancreatic Neoplasms/pathology , Simplexvirus/genetics
17.
Curr Eye Res ; 30(4): 249-57, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15823917

ABSTRACT

PURPOSE: The purpose of this study was to develop pharmacological therapeutic alternatives for ischemia-induced proliferative retinopathy. METHODS: C57BL/6J mice were placed in 76% oxygen on postnatal day 7 (P7) for 5 days. On P12 recombinant, chimeric vascular endothelial growth factor (sVEGF-R2) or sTie2 was injected intravitreally in one eye. The fellow eye received a control injection. On P17, retinal wholemounts were prepared after perfusion with fluorescein-dextran to quantify the retinopathy. RESULTS: A single intravitreal injection of sVEGF-R2 reduced pathologic vascular changes significantly (p = 0.02). No significant effect was observed after intravitreal application of sTie2 (p = 0.07), although Ang-2 was upregulated in control animals without treatment as neovascularization developed and Ang-1 was constantly transcribed (ratio PCR). CONCLUSIONS: sVEGF-R2 interferes with VEGF signaling via VEGF-R2 receptor. Thus, local application of soluble receptors for angiogenic factors is a possible therapy for proliferative retinopathy. Receptors with a wide range of ligands might prove more useful for local application than those binding few or antagonistic ligands.


Subject(s)
Angiopoietin-1/analogs & derivatives , Receptor, TIE-2/administration & dosage , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Animals , Dextrans , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescein Angiography , Fluoresceins , Injections , Mice , Mice, Inbred C57BL , Oxygen/toxicity , RNA, Messenger/metabolism , Receptor, TIE-2/metabolism , Receptors, Fc , Recombinant Fusion Proteins , Retinal Neovascularization/chemically induced , Retinal Neovascularization/pathology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vitreous Body
18.
Oncol Rep ; 13(2): 319-24, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15643518

ABSTRACT

Antibodies are among the most versatile tools used today to characterize and target molecules in cells and in biological tissues. The development of phage display libraries encoding a large repertoire of single chain antibodies, scFv, allows the rapid and efficient isolation of antibodies specific for almost any type of molecule. A great advantage of such recombinant antibodies is the possibility to functionalize them by introducing new amino acid sequences. This leads to new features that would be difficult to introduce into naturally occurring antibody molecules. This approach has been successfully applied to create molecules with new biological activities, e.g. by generating chimeric scFv antibodies carrying sequences derived from other biomolecules such as blood clotting factors or enzymes. Here, we describe a new antibody isolated from an M13 phage library that recognizes vascular endothelial growth factor receptor 2, VEGFR-2. This antibody, scFvVR-2H9 was coupled to liposomes and used to specifically target VEGFR-2-expressing human cancer cells in culture.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Liposomes , Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-2/therapeutic use , Antibody Specificity , Humans , Peptide Library , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/immunology
19.
Cancer Gene Ther ; 12(3): 257-67, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15565180

ABSTRACT

Tumor-endothelial interaction contributes to local prostate tumor growth and distant metastasis. In this communication, we designed a novel approach to target both cancer cells and their "crosstalk" with surrounding microvascular endothelium in an experimental hormone refractory human prostate cancer model. We evaluated the in vitro and in vivo synergistic and/or additive effects of a combination of conditional oncolytic adenovirus plus an adenoviral-mediated antiangiogenic therapy. In the in vitro study, we demonstrated that human umbilical vein endothelial cells (HUVEC) and human C4-2 androgen-independent (AI) prostate cancer cells, when infected with an antiangiogenic adenoviral (Ad)-Flk1-Fc vector secreting a soluble form of Flk1, showed dramatically inhibited proliferation, migration and tubular formation of HUVEC endothelial cells. C4-2 cells showed maximal growth inhibition when coinfected with Ad-Flk1-Fc and Ad-hOC-E1, a conditional replication-competent Ad vector with viral replication driven by a human osteocalcin (hOC) promoter targeting both prostate cancer epithelial and stromal cells. Using a three-dimensional (3D) coculture model, we found that targeting C4-2 cells with Ad-hOC-E1 markedly decreased tubular formation in HUVEC, as visualized by confocal microscopy. In a subcutaneous C4-2 tumor xenograft model, tumor volume was decreased by 40-60% in animals treated with Ad-Flk1-Fc or Ad-hOC-E1 plus vitamin D3 alone and by 90% in a combined treatment group, compared to untreated animals in an 8-week treatment period. Moreover, three of 10 (30%) pre-established tumors completely regressed when animals received combination therapy. Cotargeting tumor and tumor endothelium could be a promising gene therapy strategy for the treatment of both localized and metastatic human prostate cancer.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Endothelial Cells/metabolism , Genetic Therapy/methods , Neoplasm Metastasis/therapy , Neovascularization, Pathologic/prevention & control , Prostatic Neoplasms/therapy , Vascular Endothelial Growth Factor Receptor-2/therapeutic use , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenoviridae/genetics , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Cholecalciferol/administration & dosage , Cholecalciferol/therapeutic use , Endothelial Cells/pathology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Microscopy, Confocal , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Osteocalcin/administration & dosage , Osteocalcin/genetics , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Tetrazolium Salts , Thiazoles , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/genetics
20.
Wound Repair Regen ; 10(4): 222-9, 2002.
Article in English | MEDLINE | ID: mdl-12191004

ABSTRACT

Vascular endothelial growth factor (VEGF) is a potent paracrine signal for initiating angiogenesis. Although VEGF can bind to several cell surface receptors, VEGF receptor type 2 (VEGFR2, a.k.a. KDR or flk-1) is the primary receptor responsible for VEGF-induced endothelial cell proliferation. To determine whether the VEGF-VEGFR2 signaling axis has an important role in wound healing angiogenesis, we used a retrovirus to deliver a signaling-defective truncated VEGFR2 (tm VEGFR2) to block VEGF-VEGFR2-induced endothelial cell proliferation in murine wounds. We show that the retroviral construct effectively blocked phosphorylation of VEGFR2 in vitro and we were able to express the truncated receptor in murine wounds. We achieved significant reduction of angiogenesis and granulation tissue formation in murine wounds, but this did not lead to delayed wound closure. In contrast, there was a corresponding increase in wound contraction, showing that functional VEGFR2 intracellular signaling is not critical for normal closure of excisional dermal wounds. Our results show a novel relationship between wound bed vascularity and wound contraction.


Subject(s)
Genetic Vectors , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Retroviridae , Vascular Endothelial Growth Factor Receptor-2/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/physiology , Wound Healing/drug effects , Wound Healing/physiology , Wounds, Penetrating/physiopathology , Animals , Cell Line , Disease Models, Animal , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...